WO2009126789A2 - Engineering and delivery of therapeutic compositions of freshly isolated cells - Google Patents

Engineering and delivery of therapeutic compositions of freshly isolated cells Download PDF

Info

Publication number
WO2009126789A2
WO2009126789A2 PCT/US2009/040040 US2009040040W WO2009126789A2 WO 2009126789 A2 WO2009126789 A2 WO 2009126789A2 US 2009040040 W US2009040040 W US 2009040040W WO 2009126789 A2 WO2009126789 A2 WO 2009126789A2
Authority
WO
WIPO (PCT)
Prior art keywords
cells
cell
chimeric receptor
transfected
resting
Prior art date
Application number
PCT/US2009/040040
Other languages
French (fr)
Other versions
WO2009126789A3 (en
Inventor
Linhong Li
Madhusudan V. Peshwa
Original Assignee
Maxcyte, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Maxcyte, Inc. filed Critical Maxcyte, Inc.
Priority to DK09731422.3T priority Critical patent/DK2279253T3/en
Priority to JP2011504171A priority patent/JP5779090B2/en
Priority to EP09731422.3A priority patent/EP2279253B1/en
Publication of WO2009126789A2 publication Critical patent/WO2009126789A2/en
Publication of WO2009126789A3 publication Critical patent/WO2009126789A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4613Natural-killer cells [NK or NK-T]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464411Immunoglobulin superfamily
    • A61K39/464412CD19 or B4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464466Adhesion molecules, e.g. NRCAM, EpCAM or cadherins
    • A61K39/464468Mesothelin [MSLN]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N13/00Treatment of microorganisms or enzymes with electrical or wave energy, e.g. magnetism, sonic waves
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0646Natural killers cells [NK], NKT cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K2035/124Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells the cells being hematopoietic, bone marrow derived or blood cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5156Animal cells expressing foreign proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5158Antigen-pulsed cells, e.g. T-cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/599Cell markers; Cell surface determinants with CD designations not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells

Definitions

  • the present invention relates generally to the fields of cell biology, cancer biology, and immunology. More particularly, it concerns cells that have been engineered by loading them with chemical and biological agents and the resultant entities used as therapeutics in the treatment of multiple indications, including cancer.
  • Mononuclear cells encompassing for example hematopoietic stem cells, mesenchymal stem cells, endothelial progenitor cells, adipose derived stem cells, and peripheral blood mononuclear cells (PBMC), have been used in multiple applications for treatment of immune diseases and in regenerative medicine applications (Passweg
  • PBMC Peripheral blood mononuclear cells
  • Myeloid cells such as monocytes, macrophages, dendritic cells (DC)
  • DC dendritic cells
  • Lymphoid cells such as T cells, NK cells, B cells, lymphoid DC, are effective mediators of immune responses and can be further harnessed to also present antigen and stimulate na ⁇ ve and memory responses (Hong C and Park SH. Crit Rev Immunol. 2007 27(6):511-25; Martino A and Poccia F, Curr MoI Med. 2007 Nov. 7(7):658-73).
  • Antigen Presenting Cells are important sentinels for detecting and presenting antigens to the immune effector cells. They have been extensively studied for becoming the effective therapeutic agents. Factors of antigen-loading, process and presentation in the context of state of maturity of APC to engage effector cells are major concerns in the design and development of APC-based immunotherapies and vaccines. Electroloading of tumor antigens, provided in the form of nucleotides (DNA, mRNA) or proteins/lysates or multimeric antigenic formulations, allows for effective uptake and processing of antigens in freshly isolated cells without requiring efficient maturation of APC antigen uptake mechanisms.
  • DNA nucleotides
  • mRNA proteins/lysates or multimeric antigenic formulations
  • NK and T cells are important mediators of viral and tumor immune responses. They have been extensively studied for becoming the efficient therapeutic agents. Factors of efficient and specific target cell killing, procedure simplicity, cell availability and low graft versus host disease (GvHD) are the major concerns. Chimeric receptor constructs have been described which, when expressed in cells of the immune system, can enhance the immunological specific response to tumor cells and thereby bring clinical benefit to cancer patients. Expanded NK and T cells expressing a chimeric receptor can overcome HLA-type-related inhibition of the expanded NK cell killing and T cell receptor (TCR)-required T cell killing of targeted tumor cells (Imai et al. 2004).
  • TCR T cell receptor
  • Electroporation is a well recognized method for loading nucleic acids into cells to achieve transfection of the loaded cells.
  • the terminology of electroporation, electrotransfection and electroloading have been interchangablly used in the literature with emphasis on general meaning of this technology, the transgene expression and the transference of molecules into cytoplasm, respectively.
  • this method of transfecting cells is referred to as electroloading that is the method using electroporation with no transfecting reagent or biologically based packaging of the nucleic acid being loaded, such as a viral vector or viral-like particle, relying only on a transient electric field being applied to the cell to facilitate loading of the cell.
  • nucleofection is a special one involving a transfection reagent helping the transferred DNA in the cytoplasm to the nucleus.
  • Nucleofection has been reported to transfect resting T cells and NK cells using plasmid DNA treated with a proprietary nucleofection agent (Maasho et al., 2004). It was also demonstrated that resting T cell nucleofection of chimeric receptor could lead to specific target cell killing (Finney, et al, 2004). Many reports showed that nucleofection or electroloading with DNA resulted in cell toxicity to resting hematopoietic cells including lymphocytes, dendritic cells and NK cells (Trompeter et al. 2003; Li et al.
  • Loading of cells with mRNA brings several advantages, and potentially could overcome problems associated with DNA transfection, especially in respect to resting cells and cells that will be infused into a patient.
  • mRNA especially when loaded by electroloading results in minimal cell toxicity relative to loading with plasmid DNA, and this is especially true for electroloading of resting cells such as resting NK and peripheral blood mononuclear cells (PBMC) cells.
  • resting cells such as resting NK and peripheral blood mononuclear cells (PBMC) cells.
  • PBMC peripheral blood mononuclear cells
  • mRNA need not enter the cell nucleus to be expressed resting cells readily express loaded mRNA.
  • mRNA need not be transported to the nucleus, or transcribed or processed it can begin to be translated essentially immediately following entry into the cell's cytoplasm. This allows for rapid expression of the gene coded by the mRNA.
  • mRNA does not replicate or modify the heritable genetic material of cells and mRNA preparations typically contain a single protein coding sequence, which codes for the protein one wishes to have expressed in the loaded cell.
  • mRNA electroloading have been reported (Landi et al., 2007; Van De Parre et al. 2005; Rabinovich et al. 2006; Zhao et al., 2006).
  • autologous immunotherapy with resting unstimulated NK, T, PBL, and PBMC and allogeneic immunotherapy for resting unstimulated NK cells can be advantageous for treatment of cancer.
  • a method that allows removal of cells from the patient, their treatment outside the body, and their subsequent infusion in to the patient in minimal time, with minimal intervening procedure, and with minimal addition of foreign materials, particularly materials that contain replicating genetic information, or are antigenic, is desired for safety, and reasons of cost and efficiency.
  • a method that allows modification of these cells without need of extensive cell culture, more specifically without the need for the cells to undergo cell division outside the body comprises loading only of a nucleic acid that codes for only the therapeutic protein and which is not capable of replicating in the cells or modifying the genome of the cell that has been removed from the patient, and which will be returned to the patient, and which additionally does not involve the use of any other biologically or immunologically active components is desired.
  • PBMCs are removed from a patient or allogenic donor, electroloaded, a process defined herein as electrotransfection, and soon thereafter reinfused into the patient to effect enhanced biological activity in PBMC cell populations leading to enhanced therapeutic effects for treatment of patients.
  • This therapeutic approach simplifies the procedure of obtaining therapeutic compositions of cells that otherwise could only be obtained following extensive manipulation, including culture, activation, expansion and genetic modification of the expanded cells.
  • a peripheral blood mononuclear cell refers to a blood cell having a nucleus, such as a lymphocyte or monocyte.
  • An "unstimulated" PBMC refers to a PBMC that has not been activated, such as by a cytokine or antigen.
  • Certain embodiments of the present invention provide methods whereby a nucleic acid, such as a DNA or a mRNA, coding for a genetically engineered receptor is loaded into NK cells, including resting primary NK cells, by means of electroloading to provide transiently transfected NK cells that express the chimeric receptor encoded by the nucleic acid. Also disclosed are methods for transfection of NK cells with nucleic acids, such as DNAs or mRNAs, encoding for more than one chimeric receptor or a combination of a chimeric receptor with other chemical and/or biological agents.
  • nucleic acids such as DNAs or mRNAs
  • the present invention also provides for engineering NK cells by loading with a nucleic acid, such as a DNA or a mRNA, encoding for a chimeric receptor which can be used as an immunotherapeutic cell therapy for the treatment of cancer or disease of the immune system.
  • a nucleic acid such as a DNA or a mRNA
  • the present invention provides a method for transiently modifying a resting primary peripheral blood mononuclear cell (PBMC) to expresses a chimeric receptor on its surface comprising: isolating resting primary PBMCs; and electroloading the PBMCs with a nucleic acid, such as a DNA or an mRNA, encoding a chimeric receptor, whereby the electroloaded PBMCs express the chimeric receptor on its surface.
  • the PBMCs are monocytes.
  • the PBMCs are peripheral blood lymphocytes (PBLs).
  • the lymphocytes are natural killer (NK) cells, CD3+ T cells, and/or CD8+ T cells.
  • Resting PBMCs are PBMCs directly collected from peripheral blood or are thawed PBMCs that were frozen directly after collection from peripheral blood. Resting PBMCs may be cultured for a short time (e.g., less than 2 days) with or without specific stimulation of cytokines or ligands to stimulate cell activation for cell number expansion.
  • the present invention provides a method for transiently modifying a natural killer (NK) cell to expresses a chimeric receptor on its surface comprising: isolating an NK cell; and electroloading the NK cell with a nucleic acid, such as a DNA or an mRNA, encoding a chimeric receptor, whereby the electroloaded NK cell expresses the chimeric receptor on its surface.
  • a nucleic acid such as a DNA or an mRNA
  • the NK cell may be a resting NK cell or a growing NK cell line.
  • Resting NK cells are NK cells directly collected from peripheral blood or are thawed NK cells that were frozen directly after collection from peripheral blood. Resting NK cells may be cultured for a short time (e.g., less than 2 days) with or without specific stimulation of cytokines or ligands to stimulate cell activation for cell number expansion.
  • Growing NK cells are cells that have undergone cell stimulation/activation with a cytokine and ligand to activate cells to expand in cell number.
  • an “isolated” NK cell or “isolating” an NK cell refers to separating NK cells from non-NK cells such as red blood cells, monocytes, T cells, and B cells.
  • non-NK cells such as red blood cells, monocytes, T cells, and B cells.
  • methods are known for the isolation of NK cells and kits are commercially available for this purpose.
  • NK cells When NK cells are being isolated from whole blood, it may be desirable to first separate (by centrifugation, for example) the red blood cells from immune-system cells, and then to further separate the NK cells from other types of immune-system cells.
  • One approach for separating NK cells from other cells is based on the expression of different surface markers on different cell types. For example, one can select for NK cells with antibodies that bind CD56 or CD 16, which are expressed on the surface of NK cells, for positive selection.
  • isolating NK cells comprises separation of CD56+ cells from CD56- cells.
  • isolating NK cells comprises separation of CD 16+ cells from CD 16- cells.
  • isolating NK cells comprises separation of CD56+ and CD 16+ cells from CD56- and CD 16- cells.
  • Antibodies used for isolating NK cells will generally be attached to a solid support and/or magnetic particles (e.g., magnetic beads) to facilitate the separation of the captured cells from those cells that were not bound by the antibody.
  • Isolation of NK cells may also comprise depletion (i.e., negative selection) of non-NK cells from the sample by binding surface markers, such as CD 14, CD3, and/or CD 19, which are not expressed on the surface of NK cells.
  • isolating NK cells comprises depleting CD 14+, CD3+, and/or CD 19+ cells from the sample.
  • An “isolated” T cell or “isolating” a T cell refers to separating T cells from non-T cells such as red blood cells, monocytes, NK cells, and B cells.
  • non-T cells such as red blood cells, monocytes, NK cells, and B cells.
  • methods are known for the isolation of T cells and kits are commercially available for this purpose.
  • T cells When T cells are being isolated from whole blood, it may be desirable to first separate (by centrifugation, for example) the red blood cells from immune- system cells, and then to further separate the T cells from other types of immune- system cells.
  • One approach for separating T cells from other cells is based on the expression of different surface markers on different cell types. For example, one can select for T cells with antibodies that bind CD3, which is expressed on the surface of T cells, for positive selection.
  • Isolation of T cells may also comprise depletion (i.e., negative selection) of non-T cells from the sample by binding surface markers, such as CD56 and CD 16, which are not expressed on the surface of T cells.
  • PBLs may be isolated from other non-PBL PBMCs by culturing the PBMCs in a container (e.g., flask) and removing the cells that attach to the surface of the container after about 1-2 hours.
  • isolating NK cells or T cells comprises isolating peripheral blood lymphocytes (PBLs) from other cells, such as red blood cells and monocytes.
  • PBLs peripheral blood lymphocytes
  • the PBLs comprise at least 70%, 80%, 90%, 95%, 97%, 99%, or 99.5% of the cells in a composition.
  • isolating NK cells comprises isolating the NK cells from all other types of cells, including other PBLs.
  • the NK cells comprise at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 97%, 99%, or 99.5% of the cells in a composition.
  • isolating T cells comprises isolating the T cells from all other types of cells, including other PBLs.
  • the T cells comprise at least 90%, 95%, 97%, 99%, or 99.5% of the cells in a composition.
  • transient transfection and “transiently modifying” refer to the introduction of a nucleic acid molecule into a cell using a transfection process that does not usually result in the introduced nucleic acid molecule being inserted into the nuclear genome, the introduced nucleic acid molecule is, therefore, lost as the cells undergo mitosis.
  • stable transfection refers to a transfection process in which cells that have integrated the introduced nucleic acid molecule into their genome are selected. In this way, the stably transfected nucleic acid remains in the genome of the cell and its daughter cell after mitosis.
  • transiently expressing refers to the transient expression of a nucleic acid molecule in a transiently transfected cell.
  • the present invention provides a method of treating a hyperproliferative disease in a subject comprising: obtaining isolated resting primary peripheral blood mononuclear cells (PBMCs); electroloading the PBMCs with a nucleic acid, such as a DNA or an mRNA, coding for a chimeric receptor, whereby the electro-loaded PBMCs express the chimeric receptor on its surface; and administering the transfected PBMCs to the subject to treat the hyperproliferative disease in the subject.
  • the PBMCs are obtained from a donor other than the subject being treated.
  • the PBMCs are obtained from the subject with the hyperproliferative disease.
  • the present invention provides a method of treating a hyperproliferative disease in a subject comprising: obtaining isolated NK cells from a subject with a hyperproliferative disease or from a donor; electroloading the NK cells with a nucleic acid, such as a DNA or an mRNA, coding for a chimeric receptor, whereby the electroloaded NK cells express the chimeric receptor on their surfaces; and administering the transfected NK cells to the subject to treat the hyperproliferative disease in the subject.
  • the NK cells are freshly collected primary NK cells.
  • the freshly collected primary NK cells are isolated and electro-loaded immediately after they are obtained from the subject.
  • the freshly collected primary PBMCs are collected, isolated, and transfected within about 0.5 to 3 hours, 0.5 to 2 hours, or 0.5 to 1 hour.
  • the freshly collected primary PBMCs are frozen immediately after being collected from patient.
  • the PBMCs may be frozen in peripheral blood or they may be isolated and then frozen or they may be isolated, transfected and then frozen.
  • fresh primary PBMCs may be thawed cells that were frozen immediately after collection from a patient/donor or immediately after isolation following collection.
  • the transfected cells are administered to the patient within about 1 to 48 hours, 1 to 24 hours, 1 to 15 hours, 1 to 10 hours, or 1 to 5 hours from the time the cells were originally obtained from the patient or donor.
  • freshly collected cells are cells that have been collected from a subject but have not undergone cell division outside of the subject; thus, administering freshly collected cells to a subject would refer to administering cells that have not undergone cell division outside of a subject.
  • the subject is a human.
  • the hyperproliferative disease is cancer. It is contemplated that any type of cancer can be treated with the methods and compositions disclosed herein, including, for example, breast cancer, lung cancer, prostate cancer, ovarian cancer, brain cancer, liver cancer, cervical cancer, colon cancer, renal cancer, skin cancer, head & neck cancer, bone cancer, esophageal cancer, bladder cancer, uterine cancer, lymphatic cancer, stomach cancer, pancreatic cancer, testicular cancer, or leukemia.
  • the leukemia may be, for example, acute lymphocytic leukemia (ALL), acute myelogenous leukemia (AML), chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), or mantle cell lymphoma (MCL).
  • ALL acute lymphocytic leukemia
  • AML acute myelogenous leukemia
  • CLL chronic lymphocytic leukemia
  • CML chronic myelogenous leukemia
  • MCL mantle cell lymphoma
  • the transfected cells may be administered to the subject by methods well known to those of skill in the art.
  • the transfected cells may be administered by intravenous injection, intraarterial injection, intralymphatic injection, intramuscular injection, intratumoral injection, or subcutaneous injection. It is also contemplated that the transfected cells may be administered intraperitoneally.
  • the transfected cells may be administered to the subject at or near a tumor in the subject, or to a site from which a tumor has been surgically removed from the subject. However, it is not necessary that the transfected cells be administered at the tumor site to achieve a therapeutic effect. Thus, in certain embodiments the transfected cells may be administered at a site distant from the tumor site.
  • the transfected cells may be administered locally to a disease site, regionally to a disease site, or systemically. In one embodiment, the cells are administered by intravenous injection or intralymphatic injection. In another embodiment, the transfected cells are administered locally to a tumor site, such as by intratumoral injection. In some embodiments, the transfected cells are administered back in to the patient in less than 48 hours, less than 24 hours, or less than 12 hours from the time from when the peripheral blood is collected from the donor.
  • the transfected cells are administered back in to the patient within about 1 to 48 hours, 1 to 24 hours, 1 to 15 hours, 1 to 12 hours, 1 to 10 hours, or 1 to 5 hours from the time the NK cells were originally obtained from the donor.
  • the donor and the subject being treated may be the same person or different people.
  • the cells are autologous to the subject; and in other embodiments, the cells are allogenic to the subject.
  • the chimeric receptor will generally be selected based on the cell being targeted for killing.
  • the chimeric receptor is a chimeric receptor that binds a tumor antigen.
  • CD 19 is expressed on B-lineage cells. Accordingly, to kill leukemic B cells an anti-CD 19 chimeric receptor could be expressed on the surface of a PBMC, such as a NK cell, which would enhance interaction between the modified NK cells and B cells.
  • the chimeric receptor is an anti-CD 19 chimeric receptor.
  • the anti-CD 19 chimeric receptor is an anti-CD 19BBz encoding a single chain antibody conjugated with the 4-1 BB intercellular domain and the CD3 ⁇ domain.
  • the chimeric receptor is an anti-CD20, anti-FBP, anti-TAG-72, anti-CEA, anti-carboxyanhydrase IX, nati-CD171, anti-IL-13 receptor, anti-G(D)2, anti-PSMA, anti-mesothelin, anti-Lewis-Y, or anti-CD30 chimeric receptor.
  • CARs directed to these antigens may be used to treat the diseases associated with the cells that express these antigens.
  • these antigens have been associated with at least the following tumors: CD-19 (leukemia), FBP (ovarian), TAG-72 (colorectal), CEA (colorectal, breast, gastric), carboxyanhydrase IX (renal), CD171 (neuroblastoma), IL- 13 receptor (glioblastoma), G(D )2 (neuroblastoma), PSMA (prostate), mesothelin (pancreatic), Lewis-Y (myeloma), or CD30 (cutaneous lymphoma).
  • the chimeric receptor does not contain an intracellular domain. In certain embodiments, the chimeric receptor does not contain a CD28 intracellular domain.
  • the present invention provides a composition
  • a composition comprising: an electroloaded PBMC transiently expressing transgene encoded by a nucleic acid, such as a DNA or an mRNA, coding for a chimeric receptor, whereby the chimeric receptor is expressed on the surface of the electro-loaded PBMC; and a pharmaceutically acceptable carrier.
  • the PBMC is a resting PBMC.
  • the composition is frozen.
  • the chimeric receptor may be, for example, an anti-CD 19 chimeric receptor.
  • the composition does not contain a DNA, such as a DNA plasmid, encoding the chimeric receptor.
  • the composition is free or substantially free of viral vectors and viral-like particles.
  • the present invention provides a composition
  • a composition comprising: an electrotransfected NK cell transiently expressing transgene encoded by a mRNA coding for a chimeric receptor, whereby the chimeric receptor is expressed on the surface of the electrotransfected NK cell; and a pharmaceutically acceptable carrier.
  • the NK cell is a resting NK cell.
  • the composition is frozen.
  • the chimeric receptor may be, for example, an anti-CD 19 chimeric receptor.
  • the composition does not contain a DNA, such as a DNA plasmid, encoding the chimeric receptor.
  • the composition is free or substantially free of non-NK cells. In certain aspects, at least 60%, 80%, 90%, 95%, 96%, 96%, 98%, 99%, 99.5%, or 99.9% of the cells in the composition are NK cells.
  • the present invention also provides for loading antigens into PBMCs, and in particular in to antigen presenting cells (APCs), or for loading said antigens along with other chemical or biological agents that enhance effectiveness of antigen processing, antigen presentation, cell trafficking and localization, and control of immunoregulatory environment in a subject/patient, to facilitate use of freshly isolated (na ⁇ ve) and electro-loaded PBMCs as therapeutic compositions and methods for treatment of cancer and immune diseases.
  • APCs antigen presenting cells
  • the method of transfecting the cancer cells comprises use of an electroporation device as described in U.S. Patent Application Serial No. 10/225,446, incorporated herein by reference.
  • Methods and devices for electroporation are also described in, for example, published PCT Application Nos. WO 03/018751 and WO 2004/031353; US Patent Application Nos. 10/781,440, 10/080,272, and 10/675,592; and US Patent Nos. 5,720,921, 6,074,605, 6,773,669, 6,090,617, 6,485,961, 6,617,154, 5,612,207, all of which are incorporated by reference.
  • any embodiment of any of the present methods, devices, and systems may consist of or consist essentially of — rather than comprise/include/contain/have — the described steps and/or features.
  • the term “consisting of or “consisting essentially of may be substituted for any of the open-ended linking verbs recited above, in order to change the scope of a given claim from what it would otherwise be using the open-ended linking verb.
  • FIGs. 1A-1E Macromolecule loading/transfection in expanded NK cells.
  • FIG. IA shows that FITC-dextran (500k MW) could be efficiently loaded into expanded NK cells.
  • FIG. IB shows that FITC-siRNA (21-mer) could be efficiently loaded into expanded NK cells.
  • FIG. 1C shows that DNA plasmid encoding eGFP driven by CMV promoter could be transfected into 6-day expanded NK cells.
  • FIG. ID shows that mRNA encoding eGFP could be efficiently transfected into expanded NK cells with no significant viability lose.
  • FIG. IE shows proliferation of eGFP -mRNA transfected cells.
  • FIGs. 2A-2B Transfection of mRNA encoding anti-CD 19 chimeric receptor in expanded NK cells.
  • FIG. 2A shows that 65% of viable cells could express anti- CD 19 chimeric receptor.
  • FIG. 2B shows that the anti-CD 19 chimeric receptor expression was mRNA concentration dependent, and could express up to 4-5 days.
  • FIGs. 3A-3D Resting NK cells could be efficiently transfected with mRNA.
  • FIG. 3A shows NK cell phenotype after Miltenyi bead isolation.
  • FIG. 3B shows that resting NK cells could be efficiently transfected with mRNA encoding eGFP or anti- CD 19 chimeric receptor.
  • FIG. 3C shows the expression duration of the anti-CD 19 chimeric receptor in resting NK cells.
  • FIG. 3D shows a summary of viability and expression level of resting NK cells from two donors.
  • FIGs. 4A-4B Effect of CD3+ cells on OP-I killing by transfected NK cells.
  • FIG. 4A shows that CD3+ cells could be efficiently depleted with Dynal bead from 28% to 0.4%.
  • FIG. 4B shows that either depletion or no depletion of CD3+ cells from expanded NK cells did not affect the NK cell killing of OP-I cells.
  • FIGs. 5A-5C Specificity of NK cell killing.
  • FIG. 5A shows that proper transfection resulted in expression of the transgene.
  • FIG. 5B shows typical FACS data that GFP-expressed NK cells did not kill CD19-PE+ OP-I cells (3rd panels from left). Only anti-CD 19 chimeric receptor-expressed NK cells killed OP-I significantly (4th panels from left).
  • FIG. 5C shows a summary of anti-CD 19 chimeric receptor- specific OP-I killing. Electroporation alone (calcein-AM method) and GFP- transfected NK cells (antibody staining) did not kill OP-I. Anti-CD 19 chimeric receptor-expressed NK cells significantly killed OP-I .
  • FIGs. 6A-6D Duration of killing by expanded NK cells transfected with anti-
  • CD ⁇ chimeric receptor Killing was analyzed by calcein-AM method, set up at 3h (FIG. 6A), 1 day (FIG. 6B), 2 days (FIG. 6C), and 3 days (FIG. 6D) post transfection, and analyzed after 4 hours or 1 day killing.
  • FIGs. 7A-7B OP-I cell killing by NK cells from two different donors.
  • FIG. 7A shows that NK cells expanded from two different donors led to similar and significant OP-I cell killing.
  • FIG. 7B shows that resting NK cells from two different donors led to similar and significant OP-I cell killing.
  • FIGs. 8A-8B Allogeneic primary B-CLL cell killing by anti-CD 19 chimeric receptor-expressed NK cells.
  • FIG. 8A shows significantly higher cell killing of B- CLL cells by expanded NK cells with anti-CD 19 chimeric receptor expression than that by na ⁇ ve expanded NK cells.
  • B-CLL cells were from two donors.
  • FIG. 8B shows that B-CLL cells could be specifically killed by resting NK cells with anti- CD ⁇ chimeric receptor expression. The killing was efficient for at least 2 days after transfection.
  • FIGs. 9A-9D DNA uptake is toxic to resting PBL.
  • FIG. 9 A shows typical FACS analysis data.
  • FIG. 9B shows dependence of viability on time post transfection.
  • FIG. 9C shows dependence of viable cell numbers on time post transfection.
  • FIG. 9D shows dependence of expression on time post transfection.
  • FIGs. 10A-10B DNA uptake resulted in enhanced apoptosis in resting PBL. Resting PBL was transfected with 200ug/ml of plasmid DNA encoding for DsRed under CMV promoter and analyzed Id post transfection. The transfected cells were labeled with apoptosis indicator FITC-VAD-FMK (Promega, Madison, WI) following the product instruction. Apoptosis and transgene expression of the transfected cells were analyzed by FACS (FIG. 10A).
  • transfected cells without EP, DNA or caspase inhibitor Enzyme System Product, Livermore, CA
  • EP but without DNA or caspase inhibitor (+E-D-I) with EP but without DNA or caspase inhibitor (+E-D-I)
  • EP and caspase inhibitor but without DNA (+E-D+I with EP and DNA but without caspase inhibitor (+E+D-I)
  • EP, DNA and caspase inhibitor (+E+D+I were analyzed with Cell Death Detection ELISAPLUS kit (Roche, Indianapolis, IN) (FIG. 10B).
  • Caspase inhibitor could only slow the cell death of the DNA-transfected resting PBL, not stop it (data not shown).
  • -E or +E denotes the samples with or without transfection.
  • -CD 19 or +CD 19 represents the samples with or without addition of 5xlO 4 /ml (about 3% of the total cells in culture) autologous CD 19+ CLL cells. 100 IU/ml hIL-2 was added in the cell culture.
  • FIG. 12A-12D Characteristics of transfected resting PBL with ⁇ CD19 chimeric receptor.
  • FIG. 12A shows dependence of viability of transfected PBL on time post transfection.
  • FIG. 12B shows dependence of viable cell recovery on time post transfection.
  • FIG. 12C shows expansion of transfected PBL analyzed with CFSE.
  • FIG. 12D shows expansion of CD3+ cells analyzed with CFSE.
  • FIG. 13A-13D Specificity of allogeneic target cell/cell line killing by ⁇ CD19 chimeric receptor-transfected resting PBL.
  • FIG. 13A shows typical FACS analysis result of OP-I cell line killing.
  • FIG. 13B shows specific OP-I killing by 2 donors of transfected PBL.
  • FIG. 13C shows non-specific K562 cell killing by transfected PBL.
  • FIG. 13D shows specific CLL cell killing.
  • FIG. 14A-14E Specific autologous B (FIG. 14 A-C) or purified-CD19+ CLL (FIG. 14 D and E) cell killing by Resting NK cells (Id post transfection) (FIG. 14A); resting PBMC (3d post transfection) (FIG. 14B); Resting PBL (FIG. 14C, Id post transfection); resting PBMC from CLL patient (FIG. 14D, 2d post transfection) and resting CD3+ cells from CLL patient (FIG. 14E, 3d post transfection) after transfection with ⁇ CD19 chimeric receptor.
  • FIG. 15A-15D Duration of specific killing of autologous B cells by transfected resting PBL with ⁇ CD19 chimeric receptor.
  • the four-hour killing assay was performed at Id post transfection (FIG. 15A), 2d post transfection (FIG. 15B), 3d post transfection (FIG. 15C), and 7d post transfection (FIG. 15D).
  • FIG. 16 HS-Sultan lymphoblastic cell killing by CAR-transfected PBMCs in vitro.
  • FIG. 17 HS-Sultan lymphoblastic cell killing by CAR-transfected PBMCs in vivo.
  • HS-Sultan cells Ie6 cells
  • CAR-transfected PBMCs 7e6, 2.3e6, 6.7e6 and 20e6 respectively
  • FIG. 18 Cytokine-induced NK cells (LAK) transfected with mRNA encoding anti-CD 19-BBz exhibit greater cytotoxicity against HS-Sultan cells than stimulated LAK that were not transfected with mRNA encoding anti-CD 19-BBz.
  • FIG. 19 The effect of intracellular domains on CAR expression in expanded
  • FIG. 20 The effect of intracellular domains on CAR expression in expanded T cells.
  • the MFI is shown on the y axis.
  • the time post transfection is shown on the x axis.
  • FIG. 21 K562 killing by expanded T cells transfected with CARs linked to different intracellular domains.
  • FIG. 22 The CD28 intracellular domain deceases expression of CARs faster in T cells than in unstimulated resting PBMCs.
  • FIG. 23 Unstimulated resting PBMCs transfected with the ssl-28z CAR maintained K562+ cell killing at four days post transfection.
  • present invention provides methods and compositions for the prevention and treatment of diseases, such as cancer and other hyperproliferative diseases.
  • present invention provides methods for the preparation of transiently modified NK, T, PBL, and/or PBMC cells that provide previously unattained levels of cell viability following transfection, expression of a chimeric receptor that enhances specific anti-tumor activity by the modified cells, convenience and clinical applicability in autologous and allogeneic immunotherapeutic regimen, and improved precision in the transient modification, and safety in terms of risk of engineering of transfected cells.
  • the methods are applicable to a wide range of chimeric receptor constructs and therapeutic proteins.
  • chemical and/or biological agents such as for example nucleotides (DNA, mRNA, microRNA
  • an approach to development of immunotherapy products whereby unstimulated mononuclear cells obtained from peripheral blood are obtained from a patient, loaded with relevant chemical or biological agents using transient delivery of energy, such as electrical, light, sound, heat, waves, and chemical and/or biological mediation, to affect the biological activity of freshly isolated mononuclear cells, and soon thereafter reinfused into the patient to effect enhanced biological activity in specific mononuclear cell populations contained with the freshly isolated cells leading to enhanced therapeutic effects for treatment of patients.
  • This therapeutic approach may provide an alternative to the use of purified, isolated, or enriched cells, that need to be expanded/activated and transformed to impact their biological activity (potency) and thus may be preferred in multiple situations requiring medical interventions.
  • Mononuclear cells obtained from multiple sources can be effectively loaded with chemical and/or biological agents in a controlled manner using electrical energy, thereafter referred to as electroloading, to obtain desired level and duration of modulation of molecular pathways.
  • Controlled intervention of molecular pathways provides means for affecting biological activity of cells when administered back to subject / patient, thus enhancing the ability to mitigate potency and efficacy that is otherwise not provided for in the administration of unmodified, freshly isolated cells.
  • the present invention employs genetically modified natural killer cells in the treatment of hyperproliferative diseases.
  • Natural killer cells are a type of cytotoxic lymphocyte. NK cells are activated in response to interferons or macrophage-derived cytokines, and they play a major role in the rejection of tumors and cells infected by viruses. NK cells kill cancer cells and virally infected cells by releasing small cytoplasmic granules called perform and granzyme that cause the target cell to die. NK cells are characterized by their lack of the T cell receptor (CD3) and their expression of CD56 on their surface. Accordingly, these characteristics may be used to separate NK cells from other cell types. In contrast to cytotoxic T lymphocytes (CTL), NK cells do not require antigen activation and are not MHC restricted.
  • CTL cytotoxic T lymphocytes
  • Cancer cells may evade killing by NK cells because self HLA molecules on the cancer cells can bind to the killer immunoglobulin-like receptors (KIRs) and inhibit the NK cell killing.
  • KIRs killer immunoglobulin-like receptors
  • the present invention provides methods and compositions that overcome this inhibition and promotes NK cell killing of cancer cells.
  • the present invention employs genetically modified T cells in the treatment of hyperproliferative diseases.
  • T cells play a role in cell- mediated immunity.
  • TCR T cell receptor
  • Activation of CD8+ T cells and CD4+ T cells occurs through the engagement of both the T cell receptor and CD28 on the T cell by the major histocompatibility complex (MHC) peptide and B7 family members on an antigen presenting cell (APC).
  • MHC major histocompatibility complex
  • APC antigen presenting cell
  • TCR T cell receptor for antigen
  • CD28 CD28 costimulation
  • Anergic T cells show defective IL-2 production and proliferation upon restimulation via the TCR and CD28, and produce other cytokines at reduced levels.
  • Anergy may represent one mechanism of peripheral tolerance (Ramsdell et ah, 1989), and has been reported to occur in the setting of non-productive anti-tumor immunity in vivo (Staveley-O' Carroll et al, 1998).
  • Chimeric receptors generally comprise an extracellular antibody to specific antigen on the target cell surface and an activation/stimulation domain in the cytoplasm, chimeric receptor expression in NK, T, PBL, or PBMC cells directly links the NK, T, PBL, or PBMC cells to target cells and consequently allow NK or T cells to kill the target cells. Under this mechanism, the target cell killing can avoid the HLA-type -related NK cell killing inhibition and T cell receptor (TCR)- requirement for T cell-induced target cell killing.
  • the chimeric receptor is an anti-CD 19 chimeric receptor comprising a single chain antibody conjugated with the 4-1 BB intracellular domain and the CD3 ⁇ domain. Chimeric receptor molecules are described in US 2004/0038886, which is incorporated herein by reference.
  • the invention may be used in the treatment and prevention of hyperproliferative diseases including, but not limited to, cancer.
  • a hyperproliferative disease is any disease or condition which has, as part of its pathology, an abnormal increase in cell number. Included in such diseases are benign conditions such as benign prostatic hypertrophy and ovarian cysts. Also included are premalignant lesions, such as squamous hyperplasia. At the other end of the spectrum of hyperproliferative diseases are cancers.
  • a hyperproliferative disease can involve cells of any cell type. The hyperproliferative disease may or may not be associated with an increase in size of individual cells compared to normal cells.
  • hyperproliferative disease is a hyperproliferative lesion, a lesion characterized by an abnormal increase in the number of cells. This increase in the number of cells may or may not be associated with an increase in size of the lesion.
  • hyperproliferative lesions that are contemplated for treatment include benign tumors and premalignant lesions.
  • Examples include, but are not limited to, squamous cell hyperplastic lesions, premalignant epithelial lesions, psoriatic lesions, cutaneous warts, periungual warts, anogenital warts, epidermdysplasia verruciformis, intraepithelial neoplastic lesions, focal epithelial hyperplasia, conjunctival papilloma, conjunctival carcinoma, or squamous carcinoma lesion.
  • the lesion can involve cells of any cell type. Examples include keratinocytes, epithelial cells, skin cells, and mucosal cells.
  • the present invention provides methods and compositions for the treatment and prevention of cancer.
  • Cancer is one of the leading causes of death, being responsible for approximately 526,000 deaths in the United States each year.
  • the term "cancer” as used herein is defined as a tissue of uncontrolled growth or proliferation of cells, such as a tumor. Cancer develops through the accumulation of genetic alterations (Fearon and Vogelstein, 1990) and gains a growth advantage over normal surrounding cells. The genetic transformation of normal cells to neoplastic cells occurs through a series of progressive steps. Genetic progression models have been studied in some cancers, such as head and neck cancer (Califano et ah, 1996). Treatment and prevention of any type of cancer is contemplated by the present invention. The present invention also contemplates methods of prevention of cancer in a subject with a history of cancer. Examples of cancers have been listed above.
  • Electroporation Certain embodiments involve the use of electroporation to facilitate the entry of one or more nucleic acid molecules into cells of the immune system, such as natural killer (NK) cells.
  • NK natural killer
  • electroporation refers to application of an electrical current or electrical field to a cell to facilitate entry of a nucleic acid molecule into the cell.
  • electroporation may be carried out as described in U.S. Patent application serial no. 10/225,446, filed August 21, 2002, the entire disclosure of which is specifically incorporated herein by reference.
  • electroloading may be carried out as described in U.S. Patent number 5,612,207 (specifically incorporated herein by reference), U.S. Patent number 5,720,921 (specifically incorporated herein by reference), U.S. Patent number 6,074,605 (specifically incorporated herein by reference); U.S. Patent number 6,090,617 (specifically incorporated herein by reference); and U.S. patent number 6,485,961 (specifically incorporated herein by reference).
  • transfected cells for administration to a subject are contemplated by the present invention.
  • One of ordinary skill in the art would be familiar with techniques for administering cells to a subject.
  • one of ordinary skill in the art would be familiar with techniques and pharmaceutical reagents necessary for preparation of these cell prior to administration to a subject.
  • the pharmaceutical preparation will be an aqueous composition that includes the transfected cells that have been modified to express genetically engineered receptor.
  • the transfected cell is prepared using cells that have been obtained from the subject (i.e., autologous cells).
  • compositions of the present invention comprise an effective amount of a solution of the transfected cells in a pharmaceutically acceptable carrier or aqueous medium.
  • pharmaceutically acceptable carrier or aqueous medium As used herein, "pharmaceutical preparation" or
  • compositions includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like.
  • the use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the transfected cancer cells, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions.
  • preparations should meet sterility, pyrogenicity, general safety and purity standards as required by the FDA Center for Biologies.
  • the transfected cancer cells may be formulated for administration by any known route, such as by subcutaneous injection, intramuscular injection, intravascular injection, intratumoral injection, or application by any other route.
  • a person of ordinary skill in the art would be familiar with techniques for generating sterile solutions for injection or application by any other route. Determination of the number of cells to be administered will be made by one of skill in the art, and will in part be dependent on the extent and severity of cancer, and whether the transfected cells are being administered for treatment of existing cancer or prevention of cancer.
  • the preparation of the pharmaceutical composition containing the transfected cells will be known to those of skill in the art in light of the present disclosure.
  • the transfected cells may be administered with other agents that are part of the therapeutic regiment of the subject, such as other immunotherapy or chemotherapy.
  • about Ie7, Ie8, Ie9, or IeIO, or any range derivable therein, of transfected cells are administered per dose.
  • multiple doses may be administered over a period of days, weeks, months, or year.
  • a subject may receive, for example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 doses.
  • NK cells were transfected using electroporation with a DNA plasmid carrying an eGFP marker gene. One day after transfection, the viable and transfected NK cells were assayed and found to be about 50% and 30% respectively, as shown in FIG. 1C.
  • mRNA coding for eGFP was electrotransfected into expanded NK cells. About 80% of the viable cells expressed GFP. At the same time, the viability of NK cells was 67% in the untransfected control and 60% in the transfected cells. When normalized to the untransfected control cells, the viability of the transfected cells was 90%. The mRNA-transfected cells exhibited a reduced rate of cell division for one day post-transfection, but they regained the normal cell division rate subsequently. The transfected NK cells maintained approximately half of the cell number relative to control untransfected cells 4 days post-transfection (FIG.
  • An mRNA coding for an anti-CD 19 chimeric receptor was loaded into NK cells by electroloading. As shown in FIG. 2A, 65% of all the viable cells expressed the anti-CD 19 chimeric receptor as analyzed by flow cytometry one day post- transfection. The expression of the anti-CD 19 chimeric receptor increased with the increase of mRNA concentration used in the electroloading. Using an mRNA concentration of 200 ⁇ g/ml, expression of anti-CD 19 chimeric receptor was observed for 4 days following electroloading (FIG. 2B). Recovery of the anti-CD 19 chimeric receptor-modified NK cells was similar to that of cells electrotransfected with an mRNA coding for the marker gene eGFP.
  • NK cells were negatively selected attaining >90% purity with representation by minimal CD3+ cells (FIG. 3A). Electroloading of mRNA encoding for anti-CD 19 chimeric receptor resulted in expression of anti-CD 19 chimeric receptor in between 50 and 60% of electro-loaded resting NK cells (FIG. 3B) 1 day post-transfection. No significant decrease in the expression of anti-CD 19 chimeric receptor was observed in the electrotransfected NK cells 2 days post-transfection from 1 day post-transfection (FIG. 3C).
  • the viability of un-electroporated control cells and electrotransfected NK cells at 1 day post- transfection was 75% and 60% respectively, and was about 80% for transfected cells when normalized against un-electroporated control cells (FIG. 3D).
  • GFP expression of resting NK cells (>80%) and expanded NK cells (-80%, see Example 2 above) when electrotransfected with mRNA encoding for eGFP was much more efficient compared to the GFP expression of expanded NK cells (-50%) electrotransfected with DNA plasmid encoding eGFP.
  • FIG. 4A shows the similar killing curve derived from the samples either with or without CD3+ cell depletion.
  • anti-CD 19 chimeric receptor for killing leukemic cells was confirmed by comparing killing by NK cells electrotransfected using mRNA encoding for the chimeric anti-CD 19 receptor to otherwise identical NK cells electrotranfected using mRNA encoding for the marker gene eGFP or cells that received the same electroporation treatment absent any exogenous mRNA.
  • FIG. 7A summarizes cytotoxicty results of primary NK cells derived from two different donors. Anti-CD 19 chimeric receptor electrotransfected NK cells from both donors showed similar target cell lysis efficiency and kinetics.
  • target cell line OP-I Specific killing of target cell line OP-I by resting NK cells 1 day after electro loading was assayed.
  • the anti-CD 19 chimeric receptor- electro-loaded resting NK cells could efficiently kill OP-I cells.
  • E:T ratio of 8:1 about 80% of the target cells were lysed in the 4 hour co -cultivation killing assay.
  • the na ⁇ ve and GFP-expressing resting NK cells did not lyse target OP-I cells. Resting NK cells from both donors demonstrated similar lysis activity.
  • NK cells electrotransfected using mRNA encoding for the chimeric anti- CD 19 showed specific killing of allogeneic primary B-lineage leukemia cells
  • NK cells were electrotransfected using a mRNA encoding for the chimeric anti-CD 19 receptor, mRNA encoding for eGFP, and NK cells electroporated in the absence of exogenous mRNA were assayed for their ability to specifically lyse labeled B-CLL target cells.
  • a significant percentage of B-CLL cells were lysed by NK cells electrotransfected using mRNA encoding for the chimeric anti-CD 19 receptor as compared to NK cells electrotransfected using an mRNA encoding for eGFP or NK cells electroporated in the absence of exogenous mRNA.
  • Target B-CLL from two donors were used in these assays and the results were summarized in FIG. 8A.
  • NK cells electrotransfected using a mRNA encoding for the chimeric CD 19 receptor could kill B-CLL cells at least for two days after transfection (FIG. 8B) and the killing of these labeled B-CLL cells was significantly higher than by either the NK cells electrotransfected using mRNA encoding for eGFP or NK cells electroporated in the absence of exogenous mRNA.
  • FIG. 9A shows typical FACS analysis data.
  • FIGs. 9B and 9C the viability of cells electroporated in the presence of DNA was much lower in comparison to cells electroporated with mRNA or FITC-dex.
  • GFP expression was much lower in cells transfected with the DNA-GFP as compared to the mRNA-GFP (FIG. 9D).
  • DNA uptake also resulted in enhanced apoptosis in resting PBLs.
  • Resting PBLs were transfected with 200ug/ml of plasmid DNA encoding for DsRed under the control of a CMV promoter and analyzed 1 day post transfection.
  • the transfected cells were labeled with apoptosis indicator FITC-VAD-FMK (Promega, Madison, WI) following the product instructions.
  • Apoptosis and transgene expression of the transfected cells were analyzed by FACS (FIG. 10A). The percentage of apoptotic cells was more than twice as high in the transfected cells as compared to control cells (FIG. 10A).
  • transfected cells without electroporation, DNA or caspase inhibitor Enzyme System Product, Livermore, CA
  • electroporation but without DNA or caspase inhibitor (+E-D-I) with electroporation but without DNA or caspase inhibitor (+E-D-I)
  • electroporation and caspase inhibitor but without DNA (+E-D+I with electroporation and DNA but without caspase inhibitor (+E+D-I)
  • electroporation, DNA and caspase inhibitor (+E+D+I) were analyzed with Cell Death Detection ELISAPLUS kit (Roche, Indianapolis, IN) (FIG. 10B).
  • Caspase inhibitor could only slow the cell death of the DNA-transfected resting PBL, not stop it.
  • ⁇ CD19 chimeric expression was evaluated in resting PBMCs (FIG. HA), resting NK cells (FIG. 1 IB), resting PBLs (FIG. HC) from healthy donors and resting
  • FIG. 12A shows high viability of PBLs transfected with an mRNA encoding a ⁇ CD19 chimeric receptor for at least 7 days post transfection.
  • FIG. 12B shows dependence of viable cell recovery on time post transfection.
  • FIG. 12C shows expansion of transfected PBL analyzed with CFSE.
  • FIG. 12D shows expansion of CD3+ cells analyzed with
  • FIG. 13A shows typical FACS analysis result of OP-I cell line killing.
  • FIG. 13B shows specific OP-I killing by 2 donors of transfected PBLs, whereas na ⁇ ve or GFP-transfected PBLs did not specifically kill OP-I cells.
  • FIG. 13C shows non-specific K562 cell killing by transfected PBLs.
  • FIG. 13D shows specific CLL cell killing.
  • FIGs. 14A-14D show specific autologous B or purified CD 19+ CLL cell killing by resting NK cells (Id post transfection) (FIG. 14A); resting PBMCs (3d post transfection) (FIG. 14B); resting PBLs (Id post transfection) (FIG. 14C); resting PBMC from CLL patient (FIG. 14D) and resting CD3+ T cells from CLL patient (FIG. 14E) after transfection with ⁇ CD19 chimeric receptor.
  • FIGs. 15A-15D show the duration of specific killing of autologous B cells by transfected resting PBLs with ⁇ CD19 chimeric receptor. The four-hour killing assay was performed at 1 day post transfection (FIG. 15A), 2 days post transfection (FIG. 15B), 3 days post transfection (FIG. 15C), and 7 days post transfection (FIG. 15D).
  • the CD 19+ human B-lineage ALL cell line, OP-I developed at St.
  • PBMC peripheral blood mononuclear cells
  • PBMCs peripheral blood mononuclear cells
  • PBS phosphate buffered saline
  • CD3+ cells were obtained by negative purification using Miltenyi kit.
  • NK cells were negatively selected by following the protocol supplied with the Miltenyi kit (Auburn, CA) and frozen in liquid nitrogen until use.
  • Primary NK cells were expanded as previously described by Imai et al.
  • Peripheral blood mononuclear cells were cultured with thawed K562 cells that express 4- IBB ligand and membrane-bound IL-15 (K562-4-15) provided by St. Jude Children's Research Hospital and which were irradiated with 10,000 to 20,000 rad prior to culturing with NK cells.
  • Culturing of the NK cells with the target cells to allow for NK cell killing was performed in the presence of 10 IU/ml-100 U/ml IL-2, 10% FBS and antibiotics.
  • PBMCs were prepared by incubating the thawed PBMCs in a centrifuge tube for 30 minutes after thawing and collecting all cells by centrifugation.
  • PBLs were prepared by culturing the thawed PBMC in tissue culture flask for 1-2 hours and only collecting the suspended cells.
  • mRNA encoding for anti-CD 19 chimeric receptor was in vitro transcribed with T7 polymerase using an Ambion mMESSAGE mMACHINE T7 Ultra kit (Ambion, Austin, TX) with the cloned template of the pVAXl vector containing anti-CD 19 chimeric receptor. mRNA quality and quantity was analyzed by 1% agarose gel after 15 minutes denaturation at 70 0 C in mRNA denaturation buffer (Invitrogen, Carlsbad, CA) and OD260/280 measurement. The plasmid DNA encoding for eGFP on the pCI (Promega, Madison, WI) backbone under CMV promoter was used for DNA transfection.
  • the mRNA encoding for GFP was produced using the pCI-eGFP and the same Ambion kit as mentioned above.
  • FITC- dextran was purchased from Sigma (St. Luis, MO).
  • the FITC-conjugated control siRNA was purchased from Qiagen (Valencia, CA).
  • the resting NK cells in frozen medium (10% DMSO in FBS) were thawed in 37°C water bath, incubated for 0.5-lh at 37°C in the prewarmed fresh full medium (RPMI- 1640+ 10% FBS+ antibiotics) with volume of 10x that of frozen medium and ready for transfection.
  • the expanded NK cells were harvested at the indicated time points for transfection. Before transfection, the expanded NK cells were washed with MXCT EP buffer once.
  • the unstimulated resting cells were washed 2x in PBS containing 0.5% FBS and 2mM EDTA and 3x in MXCT EP buffer containing additionally 0.1% BSA. After washing, expanded NK and resting NK, PBL, PBMC.
  • T, and CD8+ cells were suspended in MXCT EP buffer, mixed with molecules to be loaded/transfected, transferred into MXCT chamber, transfected with program "Expanded-NK Cell# 3" and “Resting-NK#1" for expanded and resting NK cells respectively in MXCT GT system (Maxcyte, Gaithersburg, MD), transferred into incubation tube, incubated for 20 minutes at 37°C, and returned to the culture medium. The loading or expression efficiency was analyzed by flow cytometry.
  • NK cells were stained with goat anti-mouse (Fab) 2 polyclonal antibody conjugated with biotin (Jackson immuno Research labs, West Grove, PA) followed by peridinin chlorophyll protein- (PerCp; Becton Dickinson, San Jose, CA) labeled streptavidin staining.
  • Fab goat anti-mouse
  • PerCp peridinin chlorophyll protein-
  • the following antibodies were used for immunophenotypic characterization of expanded and transfected NK cells: anti-CD3 conjugated with fluorescein isothiocyanate (FITC), anti-CD 19 conjugated with phycoerythrin (PE), anti-CD 16- PE, and anti-CD56-PE.
  • Antibody staining was analyzed by a FACSCalibur (Becton
  • calcein-AM acetoxmethyl-calcein
  • calcein-AM Molecular Probes, Eugene, OR
  • E :T effector to target
  • the cells were resuspended in the original culture media, transferred to FACS tubes for FACS analysis at indicated time points.
  • cell killing assays described in Imai, et al. were followed. Briefly, plain target cells 10 5 cells in lOO ⁇ l were co-cultured with 100 ⁇ l of either transfected, non-transfected primary NK cells or just fresh medium at various effector to target (E:T) ratios in each well of a 96-well U-bottom tissue culture plate (Costar, Cambridge, MA). After 40Og x 5 minutes centrifugation, the cells were cultured for desired cell-killing time. The cells were harvested and co-stained with anti-CD 19- FITC and anti-CD56-PE antibodies for 20 minutes on ice. After washing in PBS, the cells were resuspended with 200 ⁇ l of PBS and analyzed by flowcytometry.
  • the cultures were performed in the absence of exogenous IL-2. FACS analysis was performed using a FACSCalibur with 15 second collection. The specific cell lysis rate (%) was calculated by 100 - Ntarget/Ncontroi x 100, where Ntarget is the number of viable target cells co-cultured with NK cells and the Ncontroi is the number of viable target cells cultured alone.
  • PBMCs Human PBMCs were electroloaded with mRNA encoding anti-CD19-BBz. Four or seven days post transfection, PBMCs (transfected or non-transfected (Na ⁇ ve)) were mixed in vitro with calceinAM-prelabeled HS-Sultan cells, a leukemia cell line that is CD 19+, at various effector:target (E:T) ratios. Cell cytotoxicity was performed by FACS 4 hours after mixing. As shown in FIG. 16, the transfected PBMCs were able to kill HS-Sultan cells at 4 days and 7 days at all E:T ratios tested, with the killing being more effective at 4 days than at 7 days. In addition, increased killing was achieved with higher E:T ratios.
  • E:T effector:target
  • PBMC 1 day post-mRNA transfection (transfected or non-transfected) with mRNA encoding anti-CD 19-BBz were mixed with HS-Sultan cells at different ratios and subcutaneously injected into mice (5 mice/group) as indicated in Table 1.
  • Table 1 Table 1.
  • Tumor volume in the mice was measured at day 0, 14, 18, and 26. As shown in FIG. 17, no measurable tumor developed in study groups 3, 4, and 5. Measurable tumor similar to that in the PBMC control was found in the study group receiving the lowest dose of CAR-expressing PBMCs (0.7e6).
  • LAK cytokine-induced NK cells
  • RNA was prepared that encodes CAR composed of an anti-mesothelin (ssl) murine single-chain Fv binding domain with the combination of 3 intracellular activation domains derived from 41BB and CD28, and the cytoplasmic portion of the TcRz chain.
  • Expanded T cells were electro loaded with ssl-myc-28-BBz, ssl-28z, ssl -BBz, or ssl-z mRNAs.
  • CAR expression decreased more quickly in the T cells transfected with mRNAs containing the CD28 intracellular domain (ssl-myc-28-BBz and ssl-28z).
  • the decrease in CAR expression correlated with a decrease in the ability of the T cells to kill cancer cells.
  • only ssl-BBz and ssl-z i.e., the mRNAs that did not have the CD28 intracellular domain
  • transfected T cells maintained K562 cell killing ability 3 days post transfection.
  • ssl-28z expression decreases faster in T cells than in PBMCs. This may be a result of the T cells doubling faster than the PBMCs.
  • the ssl-28z transfected PBMCs maintained K562+ cell killing ability at 4 days post transfection (FIG. 23).
  • compositions and methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and methods and in the steps or in the sequence of steps of the methods described herein without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents which are both chemically and physiologically related may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.
  • Interleukin-4 induces programmed cell death (apoptosis) in cases of high-risk acute lymphoblastic leukemia. Blood. 1994;83:1731-1737.

Abstract

The present invention relates to the transient modification of cells. In particular embodiments, the cells are immune systems, such as PBMC, PBL, T (CD3+ and/or CD8+) and Natural Killer (NK) cells. The modified cells provide a population of cells that express a genetically engineered chimeric receptor which can be administered to a patient therapeutically. The present invention further relates to methods that deliver mRNA coding for the chimeric receptor to unstimulated resting PBMC, PBL, T (CD3+ and/or CD8+) and NK cells and which delivers the mRNA efficiently to the transfected cells and promotes significant target cell killing.

Description

DESCRIPTION
ENGINEERING AND DELIVERY OF THERAPEUTIC COMPOSITIONS OF
FRESHLY ISOLATED CELLS
BACKGROUND OF THE INVENTION This application claims priority to United States Provisional Patent
Application Serial No. 61/043,653, filed on April 9, 2008. The entirety of the above- referenced disclosure is incorporated by reference.
1. Field of the Invention
The present invention relates generally to the fields of cell biology, cancer biology, and immunology. More particularly, it concerns cells that have been engineered by loading them with chemical and biological agents and the resultant entities used as therapeutics in the treatment of multiple indications, including cancer.
2. Description of Related Art
Mononuclear cells, encompassing for example hematopoietic stem cells, mesenchymal stem cells, endothelial progenitor cells, adipose derived stem cells, and peripheral blood mononuclear cells (PBMC), have been used in multiple applications for treatment of immune diseases and in regenerative medicine applications (Passweg
J and Tyndall A., Semin Hematol. 2007 Oct. 44(4):278-85; Le Blanc K and Ringden
O. Intern Med. 2007 Nov. 262(5):509-25; Ward et al. Catheter Cardiovasc Interv. 2007 Dec. 1 70(7):983-98; Mimeault et al., Clin Pharmacol Ther. 2007 Sep.
82(3):252-64 Epub 2007 Aug 1).
Peripheral blood mononuclear cells (PBMC) are comprised of cells of myeloid and lymphoid lineages. Myeloid cells, such as monocytes, macrophages, dendritic cells (DC), when loaded with antigens have been demonstrated to be effective antigen presenting cells for generation of tumor-antigen specific immune responses for treatment of cancer or for modulation of self-antigen specific T cells and regulatory T cells in control of autoimmunity (Gilboa E., J Clin Invest. 2007 May 117(5):1195- 203). Lymphoid cells, such as T cells, NK cells, B cells, lymphoid DC, are effective mediators of immune responses and can be further harnessed to also present antigen and stimulate naϊve and memory responses (Hong C and Park SH. Crit Rev Immunol. 2007 27(6):511-25; Martino A and Poccia F, Curr MoI Med. 2007 Nov. 7(7):658-73).
Antigen Presenting Cells (APC) are important sentinels for detecting and presenting antigens to the immune effector cells. They have been extensively studied for becoming the effective therapeutic agents. Factors of antigen-loading, process and presentation in the context of state of maturity of APC to engage effector cells are major concerns in the design and development of APC-based immunotherapies and vaccines. Electroloading of tumor antigens, provided in the form of nucleotides (DNA, mRNA) or proteins/lysates or multimeric antigenic formulations, allows for effective uptake and processing of antigens in freshly isolated cells without requiring efficient maturation of APC antigen uptake mechanisms. Further other chemical and/or biological agents can be electro-loaded into APC to affect antigen-processing, processed antigen presentation, or immuno-regulatory environment in subject/patient such that the effective biological activity of electro-loaded APC is engineered to be superior to that of naϊve freshly isolated APC. Such antigen-loading or antigen- loading combined with enhancement of biological activity for freshly isolated (naϊve) APC is a unique attribute of the composition of PBMC thus loaded allowing for rapid formulation and delivery of product to subject / patient. Such biological activity otherwise would only be imparted following processes that require elaborate cell culture, expansion, differentiation, maturation of other manipulation processes that do not lend themselves to delivery of a therapeutic composition of APC immunotherapy and vaccine products in clinically relevant timeframe for administration to subject/patient in a hospital/physician's office setting.
NK and T cells are important mediators of viral and tumor immune responses. They have been extensively studied for becoming the efficient therapeutic agents. Factors of efficient and specific target cell killing, procedure simplicity, cell availability and low graft versus host disease (GvHD) are the major concerns. Chimeric receptor constructs have been described which, when expressed in cells of the immune system, can enhance the immunological specific response to tumor cells and thereby bring clinical benefit to cancer patients. Expanded NK and T cells expressing a chimeric receptor can overcome HLA-type-related inhibition of the expanded NK cell killing and T cell receptor (TCR)-required T cell killing of targeted tumor cells (Imai et al. 2004). However, considering the simplicity of using resting NK or T cells, if the resting NK or T cells, either autologous or allogeneic, could be engineered to have both efficient and specific target cell killing, it will be the desired for the tumor therapy. Furthermore, if resting peripheral blood lymphocytes (PBL) or even peripheral blood mononuclear cells (PBMC) could be engineered to have both efficient and specific target cell killing, it will be the most desired for the tumor therapy because of procedural simplicity and cell availability. Regular retroviral vectors could not infect resting NK or T cells. Lentiviral vectors have been used to transfect resting peripheral blood lymphocytes (Simmons et al. 2006). Unfortunately, use of viral vectors entails safety and practical problems for clinical application.
Electroporation is a well recognized method for loading nucleic acids into cells to achieve transfection of the loaded cells. The terminology of electroporation, electrotransfection and electroloading have been interchangablly used in the literature with emphasis on general meaning of this technology, the transgene expression and the transference of molecules into cytoplasm, respectively. Hereinafter this method of transfecting cells is referred to as electroloading that is the method using electroporation with no transfecting reagent or biologically based packaging of the nucleic acid being loaded, such as a viral vector or viral-like particle, relying only on a transient electric field being applied to the cell to facilitate loading of the cell. Within electroporation, nucleofection is a special one involving a transfection reagent helping the transferred DNA in the cytoplasm to the nucleus. Nucleofection has been reported to transfect resting T cells and NK cells using plasmid DNA treated with a proprietary nucleofection agent (Maasho et al., 2004). It was also demonstrated that resting T cell nucleofection of chimeric receptor could lead to specific target cell killing (Finney, et al, 2004). Many reports showed that nucleofection or electroloading with DNA resulted in cell toxicity to resting hematopoietic cells including lymphocytes, dendritic cells and NK cells (Trompeter et al. 2003; Li et al. 2006; Li et al. 2001; Li et al. 1999; Landi et al., 2007; Van De Parre et al. 2005; Maasho et al. 2004; Abbott et al. 2006). Nucleofected resting NK cells or electrotransfected resting hematopoietic cells showed good transient viability and efficient transgene expression within a few hours after transfection and low viability after approximated 28 and 52 hours post-nucleofection and much decreased expression of a transgene at these times (Trompeter et al. 2003). Accordingly, this method of transient DNA transfection would not provide for a clinically useful preparation of transiently modified resting NK and T cells. Moreover, the transfection efficiency of fresh resting NK cells was about half that of growing NK cell lines.
Loading of cells with mRNA brings several advantages, and potentially could overcome problems associated with DNA transfection, especially in respect to resting cells and cells that will be infused into a patient. First, mRNA, especially when loaded by electroloading results in minimal cell toxicity relative to loading with plasmid DNA, and this is especially true for electroloading of resting cells such as resting NK and peripheral blood mononuclear cells (PBMC) cells. Also, since mRNA need not enter the cell nucleus to be expressed resting cells readily express loaded mRNA. Further, since mRNA need not be transported to the nucleus, or transcribed or processed it can begin to be translated essentially immediately following entry into the cell's cytoplasm. This allows for rapid expression of the gene coded by the mRNA. Moreover, mRNA does not replicate or modify the heritable genetic material of cells and mRNA preparations typically contain a single protein coding sequence, which codes for the protein one wishes to have expressed in the loaded cell. Various studies on mRNA electroloading have been reported (Landi et al., 2007; Van De Parre et al. 2005; Rabinovich et al. 2006; Zhao et al., 2006).
For a number of medical reasons autologous immunotherapy with resting unstimulated NK, T, PBL, and PBMC and allogeneic immunotherapy for resting unstimulated NK cells can be advantageous for treatment of cancer. In this context a method that allows removal of cells from the patient, their treatment outside the body, and their subsequent infusion in to the patient in minimal time, with minimal intervening procedure, and with minimal addition of foreign materials, particularly materials that contain replicating genetic information, or are antigenic, is desired for safety, and reasons of cost and efficiency. A method that allows modification of these cells without need of extensive cell culture, more specifically without the need for the cells to undergo cell division outside the body, comprises loading only of a nucleic acid that codes for only the therapeutic protein and which is not capable of replicating in the cells or modifying the genome of the cell that has been removed from the patient, and which will be returned to the patient, and which additionally does not involve the use of any other biologically or immunologically active components is desired.
SUMMARY OF THE INVENTION
The ability to load freshly prepared unsitmulated resting PBMCs using a method that only employs nucleic acids (DNA, mRNA, microRNA or RNAi), proteins and small molecules without requiring additional synthetic transfection reagents or viral vectors provides for a transfusion-medicine approach to development of immunotherapy products whereby PBMCs are removed from a patient or allogenic donor, electroloaded, a process defined herein as electrotransfection, and soon thereafter reinfused into the patient to effect enhanced biological activity in PBMC cell populations leading to enhanced therapeutic effects for treatment of patients. This therapeutic approach simplifies the procedure of obtaining therapeutic compositions of cells that otherwise could only be obtained following extensive manipulation, including culture, activation, expansion and genetic modification of the expanded cells. As used herein, a peripheral blood mononuclear cell (PBMC) refers to a blood cell having a nucleus, such as a lymphocyte or monocyte. An "unstimulated" PBMC refers to a PBMC that has not been activated, such as by a cytokine or antigen.
Certain embodiments of the present invention provide methods whereby a nucleic acid, such as a DNA or a mRNA, coding for a genetically engineered receptor is loaded into NK cells, including resting primary NK cells, by means of electroloading to provide transiently transfected NK cells that express the chimeric receptor encoded by the nucleic acid. Also disclosed are methods for transfection of NK cells with nucleic acids, such as DNAs or mRNAs, encoding for more than one chimeric receptor or a combination of a chimeric receptor with other chemical and/or biological agents. In some embodiments, the present invention also provides for engineering NK cells by loading with a nucleic acid, such as a DNA or a mRNA, encoding for a chimeric receptor which can be used as an immunotherapeutic cell therapy for the treatment of cancer or disease of the immune system.
In one embodiment, the present invention provides a method for transiently modifying a resting primary peripheral blood mononuclear cell (PBMC) to expresses a chimeric receptor on its surface comprising: isolating resting primary PBMCs; and electroloading the PBMCs with a nucleic acid, such as a DNA or an mRNA, encoding a chimeric receptor, whereby the electroloaded PBMCs express the chimeric receptor on its surface. In certain aspects of the invention the PBMCs are monocytes. In some aspects of the invention, the PBMCs are peripheral blood lymphocytes (PBLs). In some embodiments, the lymphocytes are natural killer (NK) cells, CD3+ T cells, and/or CD8+ T cells. Resting PBMCs are PBMCs directly collected from peripheral blood or are thawed PBMCs that were frozen directly after collection from peripheral blood. Resting PBMCs may be cultured for a short time (e.g., less than 2 days) with or without specific stimulation of cytokines or ligands to stimulate cell activation for cell number expansion.
In one embodiment, the present invention provides a method for transiently modifying a natural killer (NK) cell to expresses a chimeric receptor on its surface comprising: isolating an NK cell; and electroloading the NK cell with a nucleic acid, such as a DNA or an mRNA, encoding a chimeric receptor, whereby the electroloaded NK cell expresses the chimeric receptor on its surface.
The NK cell may be a resting NK cell or a growing NK cell line. Resting NK cells are NK cells directly collected from peripheral blood or are thawed NK cells that were frozen directly after collection from peripheral blood. Resting NK cells may be cultured for a short time (e.g., less than 2 days) with or without specific stimulation of cytokines or ligands to stimulate cell activation for cell number expansion. Growing NK cells are cells that have undergone cell stimulation/activation with a cytokine and ligand to activate cells to expand in cell number.
An "isolated" NK cell or "isolating" an NK cell refers to separating NK cells from non-NK cells such as red blood cells, monocytes, T cells, and B cells. A variety of methods are known for the isolation of NK cells and kits are commercially available for this purpose. When NK cells are being isolated from whole blood, it may be desirable to first separate (by centrifugation, for example) the red blood cells from immune-system cells, and then to further separate the NK cells from other types of immune-system cells. One approach for separating NK cells from other cells is based on the expression of different surface markers on different cell types. For example, one can select for NK cells with antibodies that bind CD56 or CD 16, which are expressed on the surface of NK cells, for positive selection. Thus, in one aspect of the invention, isolating NK cells comprises separation of CD56+ cells from CD56- cells. In another aspect of the invention, isolating NK cells comprises separation of CD 16+ cells from CD 16- cells. In a further aspect of the invention, isolating NK cells comprises separation of CD56+ and CD 16+ cells from CD56- and CD 16- cells. Antibodies used for isolating NK cells will generally be attached to a solid support and/or magnetic particles (e.g., magnetic beads) to facilitate the separation of the captured cells from those cells that were not bound by the antibody. Isolation of NK cells may also comprise depletion (i.e., negative selection) of non-NK cells from the sample by binding surface markers, such as CD 14, CD3, and/or CD 19, which are not expressed on the surface of NK cells. Thus, in one aspect of the invention, isolating NK cells comprises depleting CD 14+, CD3+, and/or CD 19+ cells from the sample.
An "isolated" T cell or "isolating" a T cell refers to separating T cells from non-T cells such as red blood cells, monocytes, NK cells, and B cells. A variety of methods are known for the isolation of T cells and kits are commercially available for this purpose. When T cells are being isolated from whole blood, it may be desirable to first separate (by centrifugation, for example) the red blood cells from immune- system cells, and then to further separate the T cells from other types of immune- system cells. One approach for separating T cells from other cells is based on the expression of different surface markers on different cell types. For example, one can select for T cells with antibodies that bind CD3, which is expressed on the surface of T cells, for positive selection. Isolation of T cells may also comprise depletion (i.e., negative selection) of non-T cells from the sample by binding surface markers, such as CD56 and CD 16, which are not expressed on the surface of T cells. PBLs, may be isolated from other non-PBL PBMCs by culturing the PBMCs in a container (e.g., flask) and removing the cells that attach to the surface of the container after about 1-2 hours.
The purity of isolated cells may be determined by, for example, fluorescence- activated cell sorting (FACS). In one embodiment of the invention, isolating NK cells or T cells comprises isolating peripheral blood lymphocytes (PBLs) from other cells, such as red blood cells and monocytes. In certain aspects, the PBLs comprise at least 70%, 80%, 90%, 95%, 97%, 99%, or 99.5% of the cells in a composition. In another embodiment of the invention, isolating NK cells comprises isolating the NK cells from all other types of cells, including other PBLs. In certain aspects, the NK cells comprise at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 97%, 99%, or 99.5% of the cells in a composition. In another embodiment of the invention, isolating T cells comprises isolating the T cells from all other types of cells, including other PBLs. In certain aspects, the T cells comprise at least 90%, 95%, 97%, 99%, or 99.5% of the cells in a composition.
The terms "transient transfection" and "transiently modifying" refer to the introduction of a nucleic acid molecule into a cell using a transfection process that does not usually result in the introduced nucleic acid molecule being inserted into the nuclear genome, the introduced nucleic acid molecule is, therefore, lost as the cells undergo mitosis. In contrast, "stable transfection" refers to a transfection process in which cells that have integrated the introduced nucleic acid molecule into their genome are selected. In this way, the stably transfected nucleic acid remains in the genome of the cell and its daughter cell after mitosis. The term "transiently expressing" refers to the transient expression of a nucleic acid molecule in a transiently transfected cell.
In one embodiment, the present invention provides a method of treating a hyperproliferative disease in a subject comprising: obtaining isolated resting primary peripheral blood mononuclear cells (PBMCs); electroloading the PBMCs with a nucleic acid, such as a DNA or an mRNA, coding for a chimeric receptor, whereby the electro-loaded PBMCs express the chimeric receptor on its surface; and administering the transfected PBMCs to the subject to treat the hyperproliferative disease in the subject. In certain embodiments, the PBMCs are obtained from a donor other than the subject being treated. In other embodiments, the PBMCs are obtained from the subject with the hyperproliferative disease.
In one embodiment, the present invention provides a method of treating a hyperproliferative disease in a subject comprising: obtaining isolated NK cells from a subject with a hyperproliferative disease or from a donor; electroloading the NK cells with a nucleic acid, such as a DNA or an mRNA, coding for a chimeric receptor, whereby the electroloaded NK cells express the chimeric receptor on their surfaces; and administering the transfected NK cells to the subject to treat the hyperproliferative disease in the subject. In certain embodiments, the NK cells are freshly collected primary NK cells. In one aspect, the freshly collected primary NK cells are isolated and electro-loaded immediately after they are obtained from the subject.
In some embodiments, the freshly collected primary PBMCs are collected, isolated, and transfected within about 0.5 to 3 hours, 0.5 to 2 hours, or 0.5 to 1 hour. In some embodiments, the freshly collected primary PBMCs are frozen immediately after being collected from patient. The PBMCs may be frozen in peripheral blood or they may be isolated and then frozen or they may be isolated, transfected and then frozen. Thus, in certain aspects of the invention, fresh primary PBMCs may be thawed cells that were frozen immediately after collection from a patient/donor or immediately after isolation following collection. In some embodiments, the transfected cells are administered to the patient within about 1 to 48 hours, 1 to 24 hours, 1 to 15 hours, 1 to 10 hours, or 1 to 5 hours from the time the cells were originally obtained from the patient or donor. In some aspects, freshly collected cells are cells that have been collected from a subject but have not undergone cell division outside of the subject; thus, administering freshly collected cells to a subject would refer to administering cells that have not undergone cell division outside of a subject.
In certain embodiments, the subject is a human. In one embodiment, the hyperproliferative disease is cancer. It is contemplated that any type of cancer can be treated with the methods and compositions disclosed herein, including, for example, breast cancer, lung cancer, prostate cancer, ovarian cancer, brain cancer, liver cancer, cervical cancer, colon cancer, renal cancer, skin cancer, head & neck cancer, bone cancer, esophageal cancer, bladder cancer, uterine cancer, lymphatic cancer, stomach cancer, pancreatic cancer, testicular cancer, or leukemia. The leukemia may be, for example, acute lymphocytic leukemia (ALL), acute myelogenous leukemia (AML), chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), or mantle cell lymphoma (MCL).
The transfected cells may be administered to the subject by methods well known to those of skill in the art. For example, the transfected cells may be administered by intravenous injection, intraarterial injection, intralymphatic injection, intramuscular injection, intratumoral injection, or subcutaneous injection. It is also contemplated that the transfected cells may be administered intraperitoneally. The transfected cells may be administered to the subject at or near a tumor in the subject, or to a site from which a tumor has been surgically removed from the subject. However, it is not necessary that the transfected cells be administered at the tumor site to achieve a therapeutic effect. Thus, in certain embodiments the transfected cells may be administered at a site distant from the tumor site. A medical practitioner will be able to determine a suitable administration route for a particular subject based, in part, on the type and location of the hyperproliferative disease. The transfected cells may be administered locally to a disease site, regionally to a disease site, or systemically. In one embodiment, the cells are administered by intravenous injection or intralymphatic injection. In another embodiment, the transfected cells are administered locally to a tumor site, such as by intratumoral injection. In some embodiments, the transfected cells are administered back in to the patient in less than 48 hours, less than 24 hours, or less than 12 hours from the time from when the peripheral blood is collected from the donor. In certain aspects of the invention, the transfected cells are administered back in to the patient within about 1 to 48 hours, 1 to 24 hours, 1 to 15 hours, 1 to 12 hours, 1 to 10 hours, or 1 to 5 hours from the time the NK cells were originally obtained from the donor. The donor and the subject being treated may be the same person or different people. Thus, in some embodiments the cells are autologous to the subject; and in other embodiments, the cells are allogenic to the subject.
The chimeric receptor will generally be selected based on the cell being targeted for killing. Thus, in one embodiment of the invention, the chimeric receptor is a chimeric receptor that binds a tumor antigen. CD 19 is expressed on B-lineage cells. Accordingly, to kill leukemic B cells an anti-CD 19 chimeric receptor could be expressed on the surface of a PBMC, such as a NK cell, which would enhance interaction between the modified NK cells and B cells. Thus, in one embodiment of the invention, the chimeric receptor is an anti-CD 19 chimeric receptor. In one aspect, the anti-CD 19 chimeric receptor is an anti-CD 19BBz encoding a single chain antibody conjugated with the 4-1 BB intercellular domain and the CD3ζ domain. In certain embodiments, the chimeric receptor is an anti-CD20, anti-FBP, anti-TAG-72, anti-CEA, anti-carboxyanhydrase IX, nati-CD171, anti-IL-13 receptor, anti-G(D)2, anti-PSMA, anti-mesothelin, anti-Lewis-Y, or anti-CD30 chimeric receptor. CARs directed to these antigens may be used to treat the diseases associated with the cells that express these antigens. For example, these antigens have been associated with at least the following tumors: CD-19 (leukemia), FBP (ovarian), TAG-72 (colorectal), CEA (colorectal, breast, gastric), carboxyanhydrase IX (renal), CD171 (neuroblastoma), IL- 13 receptor (glioblastoma), G(D )2 (neuroblastoma), PSMA (prostate), mesothelin (pancreatic), Lewis-Y (myeloma), or CD30 (cutaneous lymphoma). In certain aspects of the invention, the chimeric receptor does not contain an intracellular domain. In certain embodiments, the chimeric receptor does not contain a CD28 intracellular domain.
In another embodiment, the present invention provides a composition comprising: an electroloaded PBMC transiently expressing transgene encoded by a nucleic acid, such as a DNA or an mRNA, coding for a chimeric receptor, whereby the chimeric receptor is expressed on the surface of the electro-loaded PBMC; and a pharmaceutically acceptable carrier. In one aspect of the invention, the PBMC is a resting PBMC. In another aspect of the invention, the composition is frozen. The chimeric receptor may be, for example, an anti-CD 19 chimeric receptor. In some embodiments, the composition does not contain a DNA, such as a DNA plasmid, encoding the chimeric receptor. In certain embodiments, the composition is free or substantially free of viral vectors and viral-like particles.
In one embodiment, the present invention provides a composition comprising: an electrotransfected NK cell transiently expressing transgene encoded by a mRNA coding for a chimeric receptor, whereby the chimeric receptor is expressed on the surface of the electrotransfected NK cell; and a pharmaceutically acceptable carrier. In one aspect of the invention, the NK cell is a resting NK cell. In another aspect of the invention, the composition is frozen. The chimeric receptor may be, for example, an anti-CD 19 chimeric receptor. In some embodiments, the composition does not contain a DNA, such as a DNA plasmid, encoding the chimeric receptor. In certain embodiments, the composition is free or substantially free of non-NK cells. In certain aspects, at least 60%, 80%, 90%, 95%, 96%, 96%, 98%, 99%, 99.5%, or 99.9% of the cells in the composition are NK cells.
The present invention also provides for loading antigens into PBMCs, and in particular in to antigen presenting cells (APCs), or for loading said antigens along with other chemical or biological agents that enhance effectiveness of antigen processing, antigen presentation, cell trafficking and localization, and control of immunoregulatory environment in a subject/patient, to facilitate use of freshly isolated (naϊve) and electro-loaded PBMCs as therapeutic compositions and methods for treatment of cancer and immune diseases.
Those of skill in the art are familiar with methods of electroporation. The electroporation may be, for example, flow electroporation or static electroporation. In one embodiment, the method of transfecting the cancer cells comprises use of an electroporation device as described in U.S. Patent Application Serial No. 10/225,446, incorporated herein by reference. Methods and devices for electroporation are also described in, for example, published PCT Application Nos. WO 03/018751 and WO 2004/031353; US Patent Application Nos. 10/781,440, 10/080,272, and 10/675,592; and US Patent Nos. 5,720,921, 6,074,605, 6,773,669, 6,090,617, 6,485,961, 6,617,154, 5,612,207, all of which are incorporated by reference.
It is contemplated that any method or composition described herein can be implemented with respect to any other method or composition described herein.
Any embodiment of any of the present methods, devices, and systems may consist of or consist essentially of — rather than comprise/include/contain/have — the described steps and/or features. Thus, in any of the claims, the term "consisting of or "consisting essentially of may be substituted for any of the open-ended linking verbs recited above, in order to change the scope of a given claim from what it would otherwise be using the open-ended linking verb.
The use of the term "or" in the claims is used to mean " and/or" unless explicitly indicated to refer to alternatives only or the alternatives are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and "and/or."
Throughout this application, the term "about" is used to indicate that a value includes the standard deviation of error for the device or method being employed to determine the value. Following long-standing patent law, the words "a" and "an," when used in conjunction with the word "comprising" in the claims or specification, denotes one or more, unless specifically noted.
Other objects, features and advantages of the present invention will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating specific embodiments of the invention, are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description.
BRIEF DESCRIPTION OF THE DRAWINGS
The following drawings form part of the present specification and are included to further demonstrate certain aspects of the present invention. The invention may be better understood by reference to one or more of these drawings in combination with the detailed description of specific embodiments presented herein.
FIGs. 1A-1E. Macromolecule loading/transfection in expanded NK cells.
FIG. IA shows that FITC-dextran (500k MW) could be efficiently loaded into expanded NK cells. FIG. IB shows that FITC-siRNA (21-mer) could be efficiently loaded into expanded NK cells. FIG. 1C shows that DNA plasmid encoding eGFP driven by CMV promoter could be transfected into 6-day expanded NK cells. FIG. ID shows that mRNA encoding eGFP could be efficiently transfected into expanded NK cells with no significant viability lose. FIG. IE shows proliferation of eGFP -mRNA transfected cells.
FIGs. 2A-2B. Transfection of mRNA encoding anti-CD 19 chimeric receptor in expanded NK cells. FIG. 2A shows that 65% of viable cells could express anti- CD 19 chimeric receptor. FIG. 2B shows that the anti-CD 19 chimeric receptor expression was mRNA concentration dependent, and could express up to 4-5 days.
FIGs. 3A-3D. Resting NK cells could be efficiently transfected with mRNA.
FIG. 3A shows NK cell phenotype after Miltenyi bead isolation. FIG. 3B shows that resting NK cells could be efficiently transfected with mRNA encoding eGFP or anti- CD 19 chimeric receptor. FIG. 3C shows the expression duration of the anti-CD 19 chimeric receptor in resting NK cells. FIG. 3D shows a summary of viability and expression level of resting NK cells from two donors.
FIGs. 4A-4B. Effect of CD3+ cells on OP-I killing by transfected NK cells. FIG. 4A shows that CD3+ cells could be efficiently depleted with Dynal bead from 28% to 0.4%. FIG. 4B shows that either depletion or no depletion of CD3+ cells from expanded NK cells did not affect the NK cell killing of OP-I cells.
FIGs. 5A-5C. Specificity of NK cell killing. FIG. 5A shows that proper transfection resulted in expression of the transgene. FIG. 5B shows typical FACS data that GFP-expressed NK cells did not kill CD19-PE+ OP-I cells (3rd panels from left). Only anti-CD 19 chimeric receptor-expressed NK cells killed OP-I significantly (4th panels from left). FIG. 5C shows a summary of anti-CD 19 chimeric receptor- specific OP-I killing. Electroporation alone (calcein-AM method) and GFP- transfected NK cells (antibody staining) did not kill OP-I. Anti-CD 19 chimeric receptor-expressed NK cells significantly killed OP-I .
FIGs. 6A-6D. Duration of killing by expanded NK cells transfected with anti-
CD^ chimeric receptor. Killing was analyzed by calcein-AM method, set up at 3h (FIG. 6A), 1 day (FIG. 6B), 2 days (FIG. 6C), and 3 days (FIG. 6D) post transfection, and analyzed after 4 hours or 1 day killing.
FIGs. 7A-7B. OP-I cell killing by NK cells from two different donors. FIG. 7A shows that NK cells expanded from two different donors led to similar and significant OP-I cell killing. FIG. 7B shows that resting NK cells from two different donors led to similar and significant OP-I cell killing.
FIGs. 8A-8B. Allogeneic primary B-CLL cell killing by anti-CD 19 chimeric receptor-expressed NK cells. FIG. 8A shows significantly higher cell killing of B- CLL cells by expanded NK cells with anti-CD 19 chimeric receptor expression than that by naϊve expanded NK cells. B-CLL cells were from two donors. FIG. 8B shows that B-CLL cells could be specifically killed by resting NK cells with anti- CD^ chimeric receptor expression. The killing was efficient for at least 2 days after transfection. FIGs. 9A-9D. DNA uptake is toxic to resting PBL. Resting PBL was electroporated in the presence of plasmid DNA encoding for eGFP under CMV promoter, mRNA encoding for the eGFP and macromolecule FITC-dextran (50OkD). The viability and the expression level were monitored by trypan blue exclusion and flow cytometry analysis for up to 7 days post trans fection. FIG. 9 A shows typical FACS analysis data. FIG. 9B shows dependence of viability on time post transfection. FIG. 9C shows dependence of viable cell numbers on time post transfection. FIG. 9D shows dependence of expression on time post transfection.
FIGs. 10A-10B. DNA uptake resulted in enhanced apoptosis in resting PBL. Resting PBL was transfected with 200ug/ml of plasmid DNA encoding for DsRed under CMV promoter and analyzed Id post transfection. The transfected cells were labeled with apoptosis indicator FITC-VAD-FMK (Promega, Madison, WI) following the product instruction. Apoptosis and transgene expression of the transfected cells were analyzed by FACS (FIG. 10A). The transfected cells without EP, DNA or caspase inhibitor (Enzyme System Product, Livermore, CA) (-E-D-I), with EP but without DNA or caspase inhibitor (+E-D-I), with EP and caspase inhibitor but without DNA (+E-D+I), with EP and DNA but without caspase inhibitor (+E+D-I), and with EP, DNA and caspase inhibitor (+E+D+I) were analyzed with Cell Death Detection ELISAPLUS kit (Roche, Indianapolis, IN) (FIG. 10B). Caspase inhibitor could only slow the cell death of the DNA-transfected resting PBL, not stop it (data not shown).
FIG. 1 IA-I IE. αCD19 chimeric expression in resting PBMC (FIG. HA), resting NK cells (FIG. HB), resting PBL (FIG. HC), resting PBMC from CLL patient (FIG HD), resting T cells from CLL cells (FIG HE). -E or +E denotes the samples with or without transfection. -CD 19 or +CD 19 represents the samples with or without addition of 5xlO4/ml (about 3% of the total cells in culture) autologous CD 19+ CLL cells. 100 IU/ml hIL-2 was added in the cell culture.
FIG. 12A-12D. Characteristics of transfected resting PBL with αCD19 chimeric receptor. FIG. 12A shows dependence of viability of transfected PBL on time post transfection. FIG. 12B shows dependence of viable cell recovery on time post transfection. FIG. 12C shows expansion of transfected PBL analyzed with CFSE.
FIG. 12D shows expansion of CD3+ cells analyzed with CFSE. FIG. 13A-13D. Specificity of allogeneic target cell/cell line killing by αCD19 chimeric receptor-transfected resting PBL. FIG. 13A shows typical FACS analysis result of OP-I cell line killing. FIG. 13B shows specific OP-I killing by 2 donors of transfected PBL. FIG. 13C shows non-specific K562 cell killing by transfected PBL. FIG. 13D shows specific CLL cell killing.
FIG. 14A-14E. Specific autologous B (FIG. 14 A-C) or purified-CD19+ CLL (FIG. 14 D and E) cell killing by Resting NK cells (Id post transfection) (FIG. 14A); resting PBMC (3d post transfection) (FIG. 14B); Resting PBL (FIG. 14C, Id post transfection); resting PBMC from CLL patient (FIG. 14D, 2d post transfection) and resting CD3+ cells from CLL patient (FIG. 14E, 3d post transfection) after transfection with αCD19 chimeric receptor.
FIG. 15A-15D. Duration of specific killing of autologous B cells by transfected resting PBL with αCD19 chimeric receptor. The four-hour killing assay was performed at Id post transfection (FIG. 15A), 2d post transfection (FIG. 15B), 3d post transfection (FIG. 15C), and 7d post transfection (FIG. 15D).
FIG. 16. HS-Sultan lymphoblastic cell killing by CAR-transfected PBMCs in vitro.
FIG. 17. HS-Sultan lymphoblastic cell killing by CAR-transfected PBMCs in vivo. HS-Sultan cells (Ie6 cells) mixed with CAR-transfected PBMCs (0.7e6, 2.3e6, 6.7e6 and 20e6 respectively) were injected subcutaneously into beige SCID mice. The tumor volume was measured at indicated time points.
FIG. 18. Cytokine-induced NK cells (LAK) transfected with mRNA encoding anti-CD 19-BBz exhibit greater cytotoxicity against HS-Sultan cells than stimulated LAK that were not transfected with mRNA encoding anti-CD 19-BBz.
FIG. 19. The effect of intracellular domains on CAR expression in expanded
T cells. The percent CAR expression is shown on the y axis. The time post transfection is shown on the x axis. FIG. 20. The effect of intracellular domains on CAR expression in expanded T cells. The MFI is shown on the y axis. The time post transfection is shown on the x axis.
FIG. 21. K562 killing by expanded T cells transfected with CARs linked to different intracellular domains.
FIG. 22. The CD28 intracellular domain deceases expression of CARs faster in T cells than in unstimulated resting PBMCs.
FIG. 23. Unstimulated resting PBMCs transfected with the ssl-28z CAR maintained K562+ cell killing at four days post transfection.
DESCRIPTION OF ILLUSTRATIVE EMBODIMENTS
The present invention provides methods and compositions for the prevention and treatment of diseases, such as cancer and other hyperproliferative diseases. In certain embodiments, present invention provides methods for the preparation of transiently modified NK, T, PBL, and/or PBMC cells that provide previously unattained levels of cell viability following transfection, expression of a chimeric receptor that enhances specific anti-tumor activity by the modified cells, convenience and clinical applicability in autologous and allogeneic immunotherapeutic regimen, and improved precision in the transient modification, and safety in terms of risk of engineering of transfected cells. The methods are applicable to a wide range of chimeric receptor constructs and therapeutic proteins.
The ability to load freshly obtained resting unstimulated cells, from sources such as, for example peripheral blood, bone marrow, fat or other organ/tissue sources, using a method that employs transient energy delivery to facilitate transfer of chemical and/or biological agents, such as for example nucleotides (DNA, mRNA, microRNA or RNAi), proteins and small molecules, across a lipid bi-layer to affect the biological activity of desired cells within the freshly isolated cell population, wherein the affected biological activity is enhanced compared to the freshly isolated (non-loaded) cell compositions, and wherein the said composition of engineered cells can be safely delivered within a clinically relevant time-frame to a patient within a hospital and/or physician's office setting without requiring extensive needs for culture, expansion, differentiation or manipulation of cells, provides for unique therapeutic compositions of cells, in the context of a transfusion-medicine like approach to the development and delivery of novel therapeutic products, as effective treatment for multiple immune diseases.
Specifically, an approach to development of immunotherapy products whereby unstimulated mononuclear cells obtained from peripheral blood are obtained from a patient, loaded with relevant chemical or biological agents using transient delivery of energy, such as electrical, light, sound, heat, waves, and chemical and/or biological mediation, to affect the biological activity of freshly isolated mononuclear cells, and soon thereafter reinfused into the patient to effect enhanced biological activity in specific mononuclear cell populations contained with the freshly isolated cells leading to enhanced therapeutic effects for treatment of patients. This therapeutic approach may provide an alternative to the use of purified, isolated, or enriched cells, that need to be expanded/activated and transformed to impact their biological activity (potency) and thus may be preferred in multiple situations requiring medical interventions.
Mononuclear cells obtained from multiple sources (peripheral blood, bone marrow aspirates, lipo -aspirates, tissue-specific perfusates / isolates) can be effectively loaded with chemical and/or biological agents in a controlled manner using electrical energy, thereafter referred to as electroloading, to obtain desired level and duration of modulation of molecular pathways. Controlled intervention of molecular pathways provides means for affecting biological activity of cells when administered back to subject / patient, thus enhancing the ability to mitigate potency and efficacy that is otherwise not provided for in the administration of unmodified, freshly isolated cells.
A. Natural Killer Cells
In certain embodiments, the present invention employs genetically modified natural killer cells in the treatment of hyperproliferative diseases. Natural killer cells (NK cells) are a type of cytotoxic lymphocyte. NK cells are activated in response to interferons or macrophage-derived cytokines, and they play a major role in the rejection of tumors and cells infected by viruses. NK cells kill cancer cells and virally infected cells by releasing small cytoplasmic granules called perform and granzyme that cause the target cell to die. NK cells are characterized by their lack of the T cell receptor (CD3) and their expression of CD56 on their surface. Accordingly, these characteristics may be used to separate NK cells from other cell types. In contrast to cytotoxic T lymphocytes (CTL), NK cells do not require antigen activation and are not MHC restricted.
Cancer cells may evade killing by NK cells because self HLA molecules on the cancer cells can bind to the killer immunoglobulin-like receptors (KIRs) and inhibit the NK cell killing. The present invention provides methods and compositions that overcome this inhibition and promotes NK cell killing of cancer cells.
B. T Cells In some embodiments, the present invention employs genetically modified T cells in the treatment of hyperproliferative diseases. T cells play a role in cell- mediated immunity. One way in which T cells can be distinguished from other lymphocytes, such as B cells and NK cells, is by the presence on their cell surface of the T cell receptor (TCR). Activation of CD8+ T cells and CD4+ T cells occurs through the engagement of both the T cell receptor and CD28 on the T cell by the major histocompatibility complex (MHC) peptide and B7 family members on an antigen presenting cell (APC). Engagement of the T cell receptor for antigen (TCR) in the absence of CD28 costimulation can result in a long-term hyporesponsive state termed clonal anergy (Schwartz, 2003). Anergic T cells show defective IL-2 production and proliferation upon restimulation via the TCR and CD28, and produce other cytokines at reduced levels. Anergy may represent one mechanism of peripheral tolerance (Ramsdell et ah, 1989), and has been reported to occur in the setting of non-productive anti-tumor immunity in vivo (Staveley-O' Carroll et al, 1998).
C. Chimeric Receptors
Chimeric receptors generally comprise an extracellular antibody to specific antigen on the target cell surface and an activation/stimulation domain in the cytoplasm, chimeric receptor expression in NK, T, PBL, or PBMC cells directly links the NK, T, PBL, or PBMC cells to target cells and consequently allow NK or T cells to kill the target cells. Under this mechanism, the target cell killing can avoid the HLA-type -related NK cell killing inhibition and T cell receptor (TCR)- requirement for T cell-induced target cell killing. In one embodiment of the invention, the chimeric receptor is an anti-CD 19 chimeric receptor comprising a single chain antibody conjugated with the 4-1 BB intracellular domain and the CD3ζ domain. Chimeric receptor molecules are described in US 2004/0038886, which is incorporated herein by reference.
D. Hyperproliferative Diseases
The invention may be used in the treatment and prevention of hyperproliferative diseases including, but not limited to, cancer. A hyperproliferative disease is any disease or condition which has, as part of its pathology, an abnormal increase in cell number. Included in such diseases are benign conditions such as benign prostatic hypertrophy and ovarian cysts. Also included are premalignant lesions, such as squamous hyperplasia. At the other end of the spectrum of hyperproliferative diseases are cancers. A hyperproliferative disease can involve cells of any cell type. The hyperproliferative disease may or may not be associated with an increase in size of individual cells compared to normal cells.
Another type of hyperproliferative disease is a hyperproliferative lesion, a lesion characterized by an abnormal increase in the number of cells. This increase in the number of cells may or may not be associated with an increase in size of the lesion. Examples of hyperproliferative lesions that are contemplated for treatment include benign tumors and premalignant lesions. Examples include, but are not limited to, squamous cell hyperplastic lesions, premalignant epithelial lesions, psoriatic lesions, cutaneous warts, periungual warts, anogenital warts, epidermdysplasia verruciformis, intraepithelial neoplastic lesions, focal epithelial hyperplasia, conjunctival papilloma, conjunctival carcinoma, or squamous carcinoma lesion. The lesion can involve cells of any cell type. Examples include keratinocytes, epithelial cells, skin cells, and mucosal cells.
E. Cancer
The present invention provides methods and compositions for the treatment and prevention of cancer. Cancer is one of the leading causes of death, being responsible for approximately 526,000 deaths in the United States each year. The term "cancer" as used herein is defined as a tissue of uncontrolled growth or proliferation of cells, such as a tumor. Cancer develops through the accumulation of genetic alterations (Fearon and Vogelstein, 1990) and gains a growth advantage over normal surrounding cells. The genetic transformation of normal cells to neoplastic cells occurs through a series of progressive steps. Genetic progression models have been studied in some cancers, such as head and neck cancer (Califano et ah, 1996). Treatment and prevention of any type of cancer is contemplated by the present invention. The present invention also contemplates methods of prevention of cancer in a subject with a history of cancer. Examples of cancers have been listed above.
F. Electroporation Certain embodiments involve the use of electroporation to facilitate the entry of one or more nucleic acid molecules into cells of the immune system, such as natural killer (NK) cells.
As used herein, "electroporation" refers to application of an electrical current or electrical field to a cell to facilitate entry of a nucleic acid molecule into the cell. One of skill in the art would understand that any method and technique of electroporation is contemplated by the present invention. However, in certain embodiments of the invention, electroporation may be carried out as described in U.S. Patent application serial no. 10/225,446, filed August 21, 2002, the entire disclosure of which is specifically incorporated herein by reference.
In other embodiments of the invention, electroloading may be carried out as described in U.S. Patent number 5,612,207 (specifically incorporated herein by reference), U.S. Patent number 5,720,921 (specifically incorporated herein by reference), U.S. Patent number 6,074,605 (specifically incorporated herein by reference); U.S. Patent number 6,090,617 (specifically incorporated herein by reference); and U.S. patent number 6,485,961 (specifically incorporated herein by reference).
Other methods and devices for electroloading that may be used in the context of the present invention are also described in, for example, published PCT
Application Nos. WO 03/018751 and WO 2004/031353; US Patent Application Nos. 10/781,440, 10/080,272, and 10/675,592; and US Patent Nos. 6,773,669, 6,090,617,
6,617,154, all of which are incorporated by reference. G. Pharmaceutical Preparations 1. Formulations
Pharmaceutical preparations of transfected cells for administration to a subject are contemplated by the present invention. One of ordinary skill in the art would be familiar with techniques for administering cells to a subject. Furthermore, one of ordinary skill in the art would be familiar with techniques and pharmaceutical reagents necessary for preparation of these cell prior to administration to a subject.
In certain embodiments of the present invention, the pharmaceutical preparation will be an aqueous composition that includes the transfected cells that have been modified to express genetically engineered receptor. In certain embodiments, the transfected cell is prepared using cells that have been obtained from the subject (i.e., autologous cells).
Pharmaceutical compositions of the present invention comprise an effective amount of a solution of the transfected cells in a pharmaceutically acceptable carrier or aqueous medium. As used herein, "pharmaceutical preparation" or
"pharmaceutical composition" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the transfected cancer cells, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions. For human administration, preparations should meet sterility, pyrogenicity, general safety and purity standards as required by the FDA Center for Biologies.
The transfected cancer cells may be formulated for administration by any known route, such as by subcutaneous injection, intramuscular injection, intravascular injection, intratumoral injection, or application by any other route. A person of ordinary skill in the art would be familiar with techniques for generating sterile solutions for injection or application by any other route. Determination of the number of cells to be administered will be made by one of skill in the art, and will in part be dependent on the extent and severity of cancer, and whether the transfected cells are being administered for treatment of existing cancer or prevention of cancer. The preparation of the pharmaceutical composition containing the transfected cells will be known to those of skill in the art in light of the present disclosure. The transfected cells may be administered with other agents that are part of the therapeutic regiment of the subject, such as other immunotherapy or chemotherapy. In some embodiments, about Ie7, Ie8, Ie9, or IeIO, or any range derivable therein, of transfected cells are administered per dose. In certain aspects, multiple doses may be administered over a period of days, weeks, months, or year. A subject may receive, for example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 doses.
H. Examples The following examples are included to demonstrate certain embodiments of the invention. It should be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques discovered by the inventor to function well in the practice of the invention. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.
Example 1
Electroloading of NK cells with a DNA plasmid encoding a marker gene
NK cells were transfected using electroporation with a DNA plasmid carrying an eGFP marker gene. One day after transfection, the viable and transfected NK cells were assayed and found to be about 50% and 30% respectively, as shown in FIG. 1C.
When normalized to untransfected control cells, the viability of transfected NK cells using this plasmid DNA was about 60%. Approximately 50% of the viable NK cells expressed the eGFP marker gene. No significant change in phenotype was observed. Even though NK cells could be transfected well 1 day post transfection, the transfected NK cells lost their proliferation ability and viability because of DNA- uptake mediated cytotoxicity. DNA transfection of NK cells, therefore, would not result in meaningful clinical application. Example 2
Electroloading of expanded NK cells with mRNA coding for a marker gene
As shown in FIG. ID, mRNA coding for eGFP was electrotransfected into expanded NK cells. About 80% of the viable cells expressed GFP. At the same time, the viability of NK cells was 67% in the untransfected control and 60% in the transfected cells. When normalized to the untransfected control cells, the viability of the transfected cells was 90%. The mRNA-transfected cells exhibited a reduced rate of cell division for one day post-transfection, but they regained the normal cell division rate subsequently. The transfected NK cells maintained approximately half of the cell number relative to control untransfected cells 4 days post-transfection (FIG.
IE).
Example 3
Electroloading of expanded NK cells with mRNA coding for a chimeric receptor
An mRNA coding for an anti-CD 19 chimeric receptor was loaded into NK cells by electroloading. As shown in FIG. 2A, 65% of all the viable cells expressed the anti-CD 19 chimeric receptor as analyzed by flow cytometry one day post- transfection. The expression of the anti-CD 19 chimeric receptor increased with the increase of mRNA concentration used in the electroloading. Using an mRNA concentration of 200 μg/ml, expression of anti-CD 19 chimeric receptor was observed for 4 days following electroloading (FIG. 2B). Recovery of the anti-CD 19 chimeric receptor-modified NK cells was similar to that of cells electrotransfected with an mRNA coding for the marker gene eGFP.
Example 4
Electroloading of unstimulated resting NK cells
Freshly collected resting NK cells were isolated from a peripheral blood mononuclear cells (PBMC) population using immunoabsorbtion to magnetic beads (Miltenyi Biotec, CD56+ NK Cell Isolation Kit). NK cells were negatively selected attaining >90% purity with representation by minimal CD3+ cells (FIG. 3A). Electroloading of mRNA encoding for anti-CD 19 chimeric receptor resulted in expression of anti-CD 19 chimeric receptor in between 50 and 60% of electro-loaded resting NK cells (FIG. 3B) 1 day post-transfection. No significant decrease in the expression of anti-CD 19 chimeric receptor was observed in the electrotransfected NK cells 2 days post-transfection from 1 day post-transfection (FIG. 3C). The viability of un-electroporated control cells and electrotransfected NK cells at 1 day post- transfection was 75% and 60% respectively, and was about 80% for transfected cells when normalized against un-electroporated control cells (FIG. 3D). GFP expression of resting NK cells (>80%) and expanded NK cells (-80%, see Example 2 above) when electrotransfected with mRNA encoding for eGFP was much more efficient compared to the GFP expression of expanded NK cells (-50%) electrotransfected with DNA plasmid encoding eGFP.
Example 5
Killing of leukemia cells by NK cells electrotransfected with mRNA encoding a chimeric receptor
To assay specific killing of leukemia cells by NK cells electrotransfected by mRNA coding for a chimeric anti-CD 19 receptor the cell line OP-I was used as a target cell. Lysis was carried out in the presence or absence of CD3+ cells. Depletion of CD3+ cells in the expanded NK cell population using Dynal beads conjugated with anti-CD3 antibody (Invitrogen, Carlsbad, CA) designed for removal of CD3+ cells by positive selection following the manufacture's protocol was carried out immediately prior to electro loading with the mRNA encoding for the chimeric anti-CD 19 receptor (FIG. 4A). FIG. 4B shows the similar killing curve derived from the samples either with or without CD3+ cell depletion.
The specificity of anti-CD 19 chimeric receptor for killing leukemic cells was confirmed by comparing killing by NK cells electrotransfected using mRNA encoding for the chimeric anti-CD 19 receptor to otherwise identical NK cells electrotranfected using mRNA encoding for the marker gene eGFP or cells that received the same electroporation treatment absent any exogenous mRNA. NK cells electroporated in the absence of exogenous mRNA, NK cells electrotransfected using mRNA coding for eGFP and NK cells electrotransfected using mRNA coding for the chimeric anti- CD^ receptor exhibited similar cell viability following electro loading (FIG. 5A). NK cells electroporated in the absence of exogenous mRNA, and NK cells electrotransfected using mRNA coding for eGFP both failed to kill calcein labeled OP-I leukemia cells (FIGs. 5B and 5C). NK cells electrotransfected using mRNA encoding for the chimeric anti-CD 19 receptor when mixed with calcein labeled OP-I cells resulted in significant lysis of the target labeled OP-I cells (FIGs. 5B and 5C).
Specific lysis of labeled OP-I cells by NK cells electrotransfected using mRNA encoding for the chimeric anti-CD 19 receptor was proportional to the concentration of the mRNA encoding for the chimeric anti-CD 19 receptor. Cell killing was seen as early as 3 hours post transfection (FIG. 6A). Four hours of co- culture resulted in approximately 80% lysis of the OP-I cells at a ratio of 1 :2 (effector :target (E :T)). Overnight co-culture resulted in almost 100% lysis of the OP- 1 cells at the same 1:2 ratio. Anti-CD 19 chimeric receptor-transfected cells maintained their specific killing activity for 3 days post transfection, as shown in FIGs. 6A-6D. On day 3 post transfection, anti-CD 19 chimeric receptor-transfected cells lysed 80% and 100% of the target OP-I cells after 4 hours of co-culture at 2:1 and 4:1 E:T ratio respectively (FIG. 6D). FIG. 7A summarizes cytotoxicty results of primary NK cells derived from two different donors. Anti-CD 19 chimeric receptor electrotransfected NK cells from both donors showed similar target cell lysis efficiency and kinetics.
Specific killing of target cell line OP-I by resting NK cells 1 day after electro loading was assayed. As shown in FIG. 7B, the anti-CD 19 chimeric receptor- electro-loaded resting NK cells could efficiently kill OP-I cells. At an E:T ratio of 8:1, about 80% of the target cells were lysed in the 4 hour co -cultivation killing assay. The naϊve and GFP-expressing resting NK cells did not lyse target OP-I cells. Resting NK cells from both donors demonstrated similar lysis activity.
Example 6
Resting NK cells electrotransfected using mRNA encoding for the chimeric anti- CD 19 showed specific killing of allogeneic primary B-lineage leukemia cells
NK cells were electrotransfected using a mRNA encoding for the chimeric anti-CD 19 receptor, mRNA encoding for eGFP, and NK cells electroporated in the absence of exogenous mRNA were assayed for their ability to specifically lyse labeled B-CLL target cells. A significant percentage of B-CLL cells were lysed by NK cells electrotransfected using mRNA encoding for the chimeric anti-CD 19 receptor as compared to NK cells electrotransfected using an mRNA encoding for eGFP or NK cells electroporated in the absence of exogenous mRNA. Target B-CLL from two donors were used in these assays and the results were summarized in FIG. 8A. Resting NK cells electrotransfected using a mRNA encoding for the chimeric CD 19 receptor could kill B-CLL cells at least for two days after transfection (FIG. 8B) and the killing of these labeled B-CLL cells was significantly higher than by either the NK cells electrotransfected using mRNA encoding for eGFP or NK cells electroporated in the absence of exogenous mRNA.
Example 7
DNA Uptake is Toxic to Resting PBLs
Resting PBLs were electroporated in the presence of plasmid DNA encoding for eGFP under the control of a CMV promoter, mRNA encoding for the eGFP, and macromolecule FITC-dextran (50OkD). The viability and the expression level were monitored by trypan blue exclusion and flow cytometry analysis for up to 7 days post transfection. FIG. 9A shows typical FACS analysis data. As shown in FIGs. 9B and 9C, the viability of cells electroporated in the presence of DNA was much lower in comparison to cells electroporated with mRNA or FITC-dex. In addition, GFP expression was much lower in cells transfected with the DNA-GFP as compared to the mRNA-GFP (FIG. 9D).
DNA uptake also resulted in enhanced apoptosis in resting PBLs. Resting PBLs were transfected with 200ug/ml of plasmid DNA encoding for DsRed under the control of a CMV promoter and analyzed 1 day post transfection. The transfected cells were labeled with apoptosis indicator FITC-VAD-FMK (Promega, Madison, WI) following the product instructions. Apoptosis and transgene expression of the transfected cells were analyzed by FACS (FIG. 10A). The percentage of apoptotic cells was more than twice as high in the transfected cells as compared to control cells (FIG. 10A). The transfected cells without electroporation, DNA or caspase inhibitor (Enzyme System Product, Livermore, CA) (-E-D-I), with electroporation but without DNA or caspase inhibitor (+E-D-I), with electroporation and caspase inhibitor but without DNA (+E-D+I), with electroporation and DNA but without caspase inhibitor (+E+D-I), and with electroporation, DNA and caspase inhibitor (+E+D+I) were analyzed with Cell Death Detection ELISAPLUS kit (Roche, Indianapolis, IN) (FIG. 10B). Caspase inhibitor could only slow the cell death of the DNA-transfected resting PBL, not stop it.
Example 8
Chimeric Receptor Expression in Resting PBMCs, NK Cells, PBLs from healthy donors and resting PBMC and CD3+ T cells from CLL patient
αCD19 chimeric expression was evaluated in resting PBMCs (FIG. HA), resting NK cells (FIG. 1 IB), resting PBLs (FIG. HC) from healthy donors and resting
PBMC (FIG HD) and CD3+ cells (FIG HE) from CLL patient. FIG. 12A shows high viability of PBLs transfected with an mRNA encoding a αCD19 chimeric receptor for at least 7 days post transfection. FIG. 12B shows dependence of viable cell recovery on time post transfection. FIG. 12C shows expansion of transfected PBL analyzed with CFSE. FIG. 12D shows expansion of CD3+ cells analyzed with
CFSE.
Example 9
Specificity of Allogenic Target Cell/Cell Line Killing by αCD19 Chimeric Receptor Transfected Resting PBLs
In this study, the specificity of allogeneic target cell/cell line killing by αCD19 chimeric receptor transfected resting PBLs was evaluated. FIG. 13A shows typical FACS analysis result of OP-I cell line killing. FIG. 13B shows specific OP-I killing by 2 donors of transfected PBLs, whereas naϊve or GFP-transfected PBLs did not specifically kill OP-I cells. FIG. 13C shows non-specific K562 cell killing by transfected PBLs. FIG. 13D shows specific CLL cell killing. Example 10
Specificity of Autologous B or purified CD19+ CLL Cell Killing by αCD19
Chimeric Receptor Transfected Resting NK Cells, Resting PBMCs, Resting
PBLs from healthy donors and resting PBMC and CD3+ cells from CLL patient
FIGs. 14A-14D show specific autologous B or purified CD 19+ CLL cell killing by resting NK cells (Id post transfection) (FIG. 14A); resting PBMCs (3d post transfection) (FIG. 14B); resting PBLs (Id post transfection) (FIG. 14C); resting PBMC from CLL patient (FIG. 14D) and resting CD3+ T cells from CLL patient (FIG. 14E) after transfection with αCD19 chimeric receptor. FIGs. 15A-15D show the duration of specific killing of autologous B cells by transfected resting PBLs with αCD19 chimeric receptor. The four-hour killing assay was performed at 1 day post transfection (FIG. 15A), 2 days post transfection (FIG. 15B), 3 days post transfection (FIG. 15C), and 7 days post transfection (FIG. 15D).
Materials and Methods
Cells. The CD 19+ human B-lineage ALL cell line, OP-I (developed at St.
Jude Children's Research Hospital), and the genetically engineered myeloid leukemia cell line, K562, co-expressing 4 IBB ligand and membrane bound IL-15 (K562-4-15), also developed at St. Jude Children's Research Hospital, were maintained in RPMI- 1640 supplemented with 10% fetal bovine serum and antibiotics. Primary leukemic cells from two patients with B-CLL were obtained by directly collecting the cells in the interfacial layer after Ficoll plaque density gradient centrifugation, frozen after two rounds of PBS wash until use. Primary peripheral blood mononuclear cells (PBMC) from healthy donors were prepared from leukapheresis product purchased from BRT Laboratories, Inc. (Baltimore, MD). PBMCs were obtained directly from the interfacial layer in standard Ficoll density gradient centrifugation, washed twice with phosphate buffered saline (PBS), and frozen and stored in liquid nitrogen until use. The primary cells, whenever used, were cultured in RPMI- 1640 supplemented with 10% fetal bovine serum and antibiotics. CD3+ cells were obtained by negative purification using Miltenyi kit.
The resting NK cells were negatively selected by following the protocol supplied with the Miltenyi kit (Auburn, CA) and frozen in liquid nitrogen until use. Primary NK cells were expanded as previously described by Imai et al. Peripheral blood mononuclear cells were cultured with thawed K562 cells that express 4- IBB ligand and membrane-bound IL-15 (K562-4-15) provided by St. Jude Children's Research Hospital and which were irradiated with 10,000 to 20,000 rad prior to culturing with NK cells. Culturing of the NK cells with the target cells to allow for NK cell killing was performed in the presence of 10 IU/ml-100 U/ml IL-2, 10% FBS and antibiotics.
PBMCs were prepared by incubating the thawed PBMCs in a centrifuge tube for 30 minutes after thawing and collecting all cells by centrifugation. PBLs were prepared by culturing the thawed PBMC in tissue culture flask for 1-2 hours and only collecting the suspended cells.
Molecules for electroloading. The cloning of anti-CD 19 chimeric receptor into pVAXl (Invitrogen, Carlsbad, CA) vector was performed by digesting the parent plasmid pMSCVanti-CD19BBZ encoding a single chain antibody conjugated with the 4-1 BB intercellular domain and the CD3ζ domain (generated at St. Jude Children's Research Hospital) and the pVAXl vector with EcoR I and Xho I and Ii gated using T4 DNA ligase. mRNA encoding for anti-CD 19 chimeric receptor was in vitro transcribed with T7 polymerase using an Ambion mMESSAGE mMACHINE T7 Ultra kit (Ambion, Austin, TX) with the cloned template of the pVAXl vector containing anti-CD 19 chimeric receptor. mRNA quality and quantity was analyzed by 1% agarose gel after 15 minutes denaturation at 700C in mRNA denaturation buffer (Invitrogen, Carlsbad, CA) and OD260/280 measurement. The plasmid DNA encoding for eGFP on the pCI (Promega, Madison, WI) backbone under CMV promoter was used for DNA transfection. The mRNA encoding for GFP was produced using the pCI-eGFP and the same Ambion kit as mentioned above. FITC- dextran was purchased from Sigma (St. Luis, MO). The FITC-conjugated control siRNA was purchased from Qiagen (Valencia, CA).
Transfection. The resting NK cells in frozen medium (10% DMSO in FBS) were thawed in 37°C water bath, incubated for 0.5-lh at 37°C in the prewarmed fresh full medium (RPMI- 1640+ 10% FBS+ antibiotics) with volume of 10x that of frozen medium and ready for transfection. The expanded NK cells were harvested at the indicated time points for transfection. Before transfection, the expanded NK cells were washed with MXCT EP buffer once. The unstimulated resting cells were washed 2x in PBS containing 0.5% FBS and 2mM EDTA and 3x in MXCT EP buffer containing additionally 0.1% BSA. After washing, expanded NK and resting NK, PBL, PBMC. T, and CD8+ cells were suspended in MXCT EP buffer, mixed with molecules to be loaded/transfected, transferred into MXCT chamber, transfected with program "Expanded-NK Cell# 3" and "Resting-NK#1" for expanded and resting NK cells respectively in MXCT GT system (Maxcyte, Gaithersburg, MD), transferred into incubation tube, incubated for 20 minutes at 37°C, and returned to the culture medium. The loading or expression efficiency was analyzed by flow cytometry.
Detection of the expression of chimeric receptor and immunophenotyping. The transfected NK cells were stained with goat anti-mouse (Fab)2 polyclonal antibody conjugated with biotin (Jackson immuno Research labs, West Grove, PA) followed by peridinin chlorophyll protein- (PerCp; Becton Dickinson, San Jose, CA) labeled streptavidin staining. The positive cells was gated according to the background cells with goat biotin-conjugated IgG followed by streptavidin-PerCp .
The following antibodies were used for immunophenotypic characterization of expanded and transfected NK cells: anti-CD3 conjugated with fluorescein isothiocyanate (FITC), anti-CD 19 conjugated with phycoerythrin (PE), anti-CD 16- PE, and anti-CD56-PE. Antibody staining was analyzed by a FACSCalibur (Becton
Dickinson).
Cell Killing Assays. To facilitate the large number of cell killing studies, a cell killing assay was developed based on acetoxmethyl-calcein (calcein-AM, Molecular Probes, Eugene, OR) staining and flow cytometry. Briefly, calcein-AM pre-labeled target cells (100 μl) were co-cultured with 100 μl of either transfected, non-transfected primary NK cells or just fresh medium at various effector to target (E :T) ratios in each well of a 96-well U-bottom tissue culture plate (Costar, Cambridge, MA). The 96-well plate was centrifuged at 40Og for 5 minutes prior to cell culture at a 37°C, 5% CO2 incubator. The cells were resuspended in the original culture media, transferred to FACS tubes for FACS analysis at indicated time points. In some studies, cell killing assays described in Imai, et al. were followed. Briefly, plain target cells 105 cells in lOOμl were co-cultured with 100 μl of either transfected, non-transfected primary NK cells or just fresh medium at various effector to target (E:T) ratios in each well of a 96-well U-bottom tissue culture plate (Costar, Cambridge, MA). After 40Og x 5 minutes centrifugation, the cells were cultured for desired cell-killing time. The cells were harvested and co-stained with anti-CD 19- FITC and anti-CD56-PE antibodies for 20 minutes on ice. After washing in PBS, the cells were resuspended with 200 μl of PBS and analyzed by flowcytometry.
The cultures were performed in the absence of exogenous IL-2. FACS analysis was performed using a FACSCalibur with 15 second collection. The specific cell lysis rate (%) was calculated by 100 - Ntarget/Ncontroi x 100, where Ntarget is the number of viable target cells co-cultured with NK cells and the Ncontroi is the number of viable target cells cultured alone.
Example 11
C AR-Expressing PBMCs Kill HS-Sultan Cells In Vitro and In Vivo
Human PBMCs were electroloaded with mRNA encoding anti-CD19-BBz. Four or seven days post transfection, PBMCs (transfected or non-transfected (Naϊve)) were mixed in vitro with calceinAM-prelabeled HS-Sultan cells, a leukemia cell line that is CD 19+, at various effector:target (E:T) ratios. Cell cytotoxicity was performed by FACS 4 hours after mixing. As shown in FIG. 16, the transfected PBMCs were able to kill HS-Sultan cells at 4 days and 7 days at all E:T ratios tested, with the killing being more effective at 4 days than at 7 days. In addition, increased killing was achieved with higher E:T ratios.
To demonstrate that CAR-expressing PBMCs could kill tumor cells in vivo, a HS-Sultan subcutaneous co-mixing model in Beige SCID mice was used. PBMC 1 day post-mRNA transfection (transfected or non-transfected) with mRNA encoding anti-CD 19-BBz were mixed with HS-Sultan cells at different ratios and subcutaneously injected into mice (5 mice/group) as indicated in Table 1. Table 1.
Figure imgf000034_0001
Tumor volume in the mice was measured at day 0, 14, 18, and 26. As shown in FIG. 17, no measurable tumor developed in study groups 3, 4, and 5. Measurable tumor similar to that in the PBMC control was found in the study group receiving the lowest dose of CAR-expressing PBMCs (0.7e6).
In a further in vitro study, cytokine-induced NK cells (LAK) were transfected with mPvNA encoding anti-CD 19-BBz and mixed with HS-Sultan cells for a cytotoxicity study. LAK cells are NK cells that have been stimulated to be cytotoxic to tumor cells by Interleukin-2. As shown in FIG. 18, while control LAK cells (conventional) were cytotoxic against HS-Sultan cells, the transfected LAK cells (Engineered LAK cells) were significantly more cytotoxic.
Example 12
Effect of Intracellular Domains on CAR Expression
The effect of intracellular domains in the chimeric antigen receptor was evaluated with the following four anti-mesothelin CARs: ssl-28-BBz, ssl-28z, ssl- BBz, or ss 1 -z. RNA was prepared that encodes CAR composed of an anti-mesothelin (ssl) murine single-chain Fv binding domain with the combination of 3 intracellular activation domains derived from 41BB and CD28, and the cytoplasmic portion of the TcRz chain.
Expanded T cells were electro loaded with ssl-myc-28-BBz, ssl-28z, ssl -BBz, or ssl-z mRNAs. As shown in FIGs. 19 and 20, CAR expression decreased more quickly in the T cells transfected with mRNAs containing the CD28 intracellular domain (ssl-myc-28-BBz and ssl-28z). The decrease in CAR expression correlated with a decrease in the ability of the T cells to kill cancer cells. As shown in FIG. 21, only ssl-BBz and ssl-z (i.e., the mRNAs that did not have the CD28 intracellular domain) transfected T cells maintained K562 cell killing ability 3 days post transfection.
The expression of ss l-28z was compared in PBMCs and expanded T cells. As shown in FIG. 22, ssl-28z expression decreases faster in T cells than in PBMCs. This may be a result of the T cells doubling faster than the PBMCs. The ssl-28z transfected PBMCs maintained K562+ cell killing ability at 4 days post transfection (FIG. 23).
All of the compositions and methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and methods and in the steps or in the sequence of steps of the methods described herein without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents which are both chemically and physiologically related may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.
REFERENCES
The following references, to the extent that they provide exemplary procedural or other details supplementary to those set forth herein, are specifically incorporated herein by reference.
U.S. Patent 4,683,202
U.S. Patent 5,612,207
U.S. Patent 5,720,921
U.S. Patent 5,925,565
U.S. Patent 5,928,906
U.S. Patent 5,935,819
U.S. Patent 6,074,605
U.S. Patent 6,090,617
U.S. Patent 6,090,617
U.S. Patent 6,485,961
U.S. Patent 6,617,154
U.S. Patent 6,773,669
U.S. Application Serial No. 10/781,440
U.S. Application Serial No. 10/675,592
U.S. Application Serial No. 10/399,364
U.S. Application Serial No. 10/225,446
U.S. Application Serial No. 10/225,446
U.S. Application Serial No. 10/080,272
Almendro et al., J. Immunol., 157(12):5411-5421, 1996.
Angel et al., Mol. Cell. Biol., 7:2256, 1987a.
Angel et al, Cell, 49:729, 1987b.
Atchison and Perry, Cell, 46:253, 1986.
Atchison and Perry, Cell, 48:121, 1987.
Ausubel et al., In: Current Protocols in Molecular Biology, John, Wiley & Sons, Inc,
New York, 1996.
Banerji et al, Cell, 27(2 Pt l):299-308, 1981. Banerji et al, Cell, 33(3):729-740, 1983. Berkhout et al, Cell, 59:273-282, 1989.
Biagi et al, Hum. Gen Ther., 14(6):545-559, 2003.
Blanar et al, EMBO J., 8:1139, 1989.
Bodine and Ley, EMBO J., 6:2997, 1987.
Boshart et al, Cell, 41 :521, 1985.
Bosze ^ α/., £7kffiOJ., 5(7):1615-1623, 1986.
Braddock et al, Cell, 58:269, 1989.
Bulla and Siddiqui, J. Virol, 62:1437, 1986.
Califano et al, Cancer Res., 56(l l):2488-2492, 1996.
Campbell and Villarreal, MoI Cell. Biol, 8:1993, 1988.
Campere and Tilghman, Genes andDev., 3:537, 1989.
Campo et al, Nature, 303:77, 1983.
Carbonelli et al, FEMS Microbiol. Lett., 177(l):75-82, 1999.
Celander and Haseltine, J. Virology, 61 :269, 1987.
Cdandev et al, J. Virology, 62:1314, 1988.
Chandler et al, Cell, 33:489, 1983.
Chandler et al, Proc. Natl. Acad. Sci. USA, 94(8):3596-601, 1997.
Chang et al, MoI Cell. Biol, 9:2153, 1989.
Chatterjee et al, Proc. Natl. Acad. Sci. USA, 86:9114, 1989.
Chen et al, Oncogene, 8:2159-66, 1993.
Choi et al, Cell, 53:519, 1988.
Cocea, Biotechniques, 23(5):814-816, 1997.
Cohen et al, J. Cell. Physiol, 5:75, 1987.
Connor et al, Int. J. Gynecol. Cancer, 3(2):103-109, 1993.
Connor et al, J. Exp. Med., 177:1127-1134, 1993.
Costa et al, MoI Cell. Biol, 8:81, 1988.
Cripe et al, EMBO J., 6:3745, 1987.
Culotta and Hamer, MoI Cell. Biol, 9:1376, 1989.
Dandolo et al, J. Virology, 47:55-64, 1983.
De Jager et al, Semin. Nucl Med, 23(2):165-179, 1993.
De Villiers et al, Nature, 312(5991):242-246, 1984.
Deschamps et al, Science, 230:1174-1177, 1985.
Dilloo et al, Blood, 90(5):1927-1933, 1997.
Doolittle and Ben-Zeev, Methods MoI Biol, 109:215-237, 1999. Dranoff et al, Proc. Natl. Acad. Sci. USA, 90:3539-3543, 1993.
Edbrooke et al., MoI. Cell. Biol., 9:1908, 1989.
Edlund et al, Science, 230:912-916, 1985.
Fearon and Vogelstein, Cell, 61(5):759-767, 1990.
Feng and Holland, Nature, 334:6178, 1988.
Firak and Subramanian, MoI. Cell. Biol., 6:3667, 1986.
Foecking and Hofstetter, Gene, 45(1): 101-105, 1986.
Fujita et al, Cell, 49:357, 1987.
Gabriel and Teissie, Biophys. J, 76(4):2158-2165, 1999.
Gansbacher et al., Cancer ites.,50(24):7820-7825, 1990.
Gansbacher et al, J. Exp. Med., 172:1217-1224, 1990.
Gilles et al, Cell, 33:717, 1983.
Gloss et al, EMBO J., 6:3735, 1987.
Godbout et al, MoI Cell. Biol, 8:1169, 1988.
Goodbourn and Maniatis, Proc. Natl. Acad. Sci. USA, 85:1447, 1988.
Goodbourn et al, Cell, 45:601, 1986.
Greene et al, Immunology Today, 10:272, 1989
Grosschedl and Baltimore, Cell, 41 :885, 1985.
Gulbis and Galand, Hum. Pathol, 24(12):1271-1285, 1993.
Harding, Eur. J. Immunol, 22(7):1865-1869, 1992.
Haslinger and Karin, Proc. Natl. Acad. Sci. USA, 82:8572, 1985.
Hauber and Cullen, J. Virology, 62:673, 1988.
Hen et al, Nature, 321 :249, 1986.
Hensel et al, Lymphokine Res., 8:347, 1989.
Herr and Clarke, Cell, 45:461, 1986.
Hibino et al, Biophys. J, 64(6):1789-1800, 1993.
Hirochika ^ α/., J. Virol, 61 :2599, 1987. ϋivsch et al, MoI Cell. Biol, 10:1959, 1990.
Holbrook e^/., Virology, 157:211, 1987.
Horlick and Benfϊeld, MoI Cell. Biol, 9:2396, 1989.
Huang et al, Cell, 27:245, 1981.
Hug et al, MoI Cell. Biol, 8:3065, 1988.
Hwang et al, MoI Cell. Biol, 10:585, 1990.
Imagawa et al, Cell, 51 :251, 1987. Imbra and Karin, Nature, 323:555, 1986.
Imler et al., Mol. Cell. Biol, 7:2558, 1987.
Imperiale and Nevins, MoI. Cell. Biol, 4:875, 1984.
Jain et al, Ann. Surg. Oncology, 10:810-820, 2003.
Jakobovits et al, MoI Cell. Biol, 8:2555, 1988.
Jameel and Siddiqui, MoI Cell. Biol, 6:710, 1986.
Jaynes et al, MoI Cell. Biol, 8:62, 1988.
Johnson et al, MoI Cell. Biol, 9:3393, 1989.
Kadesch and Berg, MoI Cell. Biol, 6:2593, 1986.
Rakorin et α/., Biophys. Chem., 58(1-2): 109-116, 1996.
Karin et al, MoI Cell. Biol, 7:606, 1987.
Karp et al, J. Immunol, 150(3):896-908, 1993.
Kasaian et al, Immunity, 16:559-569, 2002.
Katinka et al, Cell, 20:393, 1980.
Kawamoto et al, MoI Cell. Biol, 8:267, 1988.
Kiledjian et al, MoI Cell. Biol, 8:145, 1988.
Kim et al, J. Virol, 76(4)1892-1903, 2002.
Kimura et al, Arch. Otolaryngol Head Neck Surg. , 129:1181-1185, 2003.
Kishida et al, Gene Therapy, 8:1234-1240, 2001.
Kishida et al, MoI Ther., 8(4):552-558, 2003.
Klamut et al, MoI Cell. Biol, 10:193, 1990.
Rnutson and Yee, Anal. Biochem., 164(l):44-52, 1987.
Koch et al, MoI Cell. Biol, 9:303, 1989.
Kotnik et al, Bioenerg., 43:281-291; 45:3-16, 1998.
Rraus et al FEBS Lett., 428(3):165-170, 1998.
Rriegler and Botchan, In: Eukaryotic Viral Vectors, Gluzman (Ed.), Cold Spring
Harbor: Cold Spring Harbor Laboratory, NY, 1982. Rriegler and Botchan, MoI Cell. Biol, 3:325, 1983. Rriegler et al, Cell, 38:483, 1984. Rriegler et al, Cell, 53:45, 1988. Ruhl et al, Cell, 50:1057, 1987. Runz et al, Nucl Acids Res., 17:1121, 1989. Lareyre et al, J. Biol. Chem., 274(12):8282-8290, 1999. Larsen et al, Proc Natl. Acad. Sci. USA., 83:8283, 1986. Laspia et al, Cell, 59:283, 1989.
Laήmev et al, MoI Cell. Biol., 10:760, 1990.
Lee et αl, DNA Cell Biol., 16(11):1267-1275, 1997.
Lee et αl, Nature, 294:228, 1981.
Lee et al, Nucleic Acids Res., 12:4191-206, 1984.
Levenson et al, Hum. Gene Ther., 9(8):1233-1236, 1998.
Levinson et al, Nature, 295:79, 1982.
Li et al, In: Am. Soc. Gene Therapy, Abstract, 5th Annual Meeting, 2002.
Li et al, J. Leukoc. Biol, 56(5):616-624, 1994.
Lin et al, MoI Cell. Biol, 10:850, 1990.
Luria et al, EMBO J., 6:3307, 1987.
Lusky and Botchan, Proc. Natl. Acad. Sci. USA, 83:3609, 1986.
Lusky et al, MoI Cell. Biol, 3:1108, 1983.
Ma et al, J. Immunol, 171(2):608-615, 2003.
Macejak and Sarnow, Nature, 353:90-94, 1991.
Majors and Varmus, Proc. Natl. Acad. Sci. USA, 80:5866, 1983.
McNeall et al, Gene, 76:81, 1989.
Mellman and Steinman, Cell, 106(3):255-258, 2001.
Miksicek et al, Cell, 46:203, 1986.
Mir, Bioelectrochem., 53:1-10, 2000.
Mordacq and Linzer, Genes andDev., 3:760, 1989.
Moreau et al, Nucl Acids Res., 9:6047, 1981.
Muesing et al, Cell, 48:691, 1987.
Nakamura et al, In: Handbook of Experimental Immunology (4th Ed.), Weir et al
(Eds.), 1 :27, Blackwell Scientific Publ, Oxford, 1987. Neumann et al, Proc. Natl. Acad. Sci. USA, 96(16):9345-9350, 1999. Ng et al, Nuc. Acids Res., 17:601, 1989. Nomoto et al, Gene, 236(2):259-271, 1999. Ondck et al, EMBO J, 6:1017, 1987. Ornitz et al, MoI Cell. Biol, 7:3466, 1987. Palmiter et al, Nature, 300:611, 1982. PCT Appln. WO 03/018751 PCT Appln. WO 03/018751 PCT Appln. WO 04/031353 Pech et al., Mol. Cell. Biol, 9:396, 1989.
Pelletier and Sonenberg, Nature, 334(6180):320-325, 1988.
Perez-Stable and Constantini, MoI. Cell. Biol, 10:1116, 1990.
Picard and Schaffher, Nature, 307:83, 1984.
VmkQxi et al, Genes and Dev., 1 :268, 1987.
Ponta et al, Proc. Natl. Acad. Sci. USA, 82:1020, 1985.
Porton et al, MoI Cell. Biol, 10:1076, 1990.
Queen and Baltimore, Cell, 35:741, 1983.
Quinn et al, MoI Cell. Biol, 9:4713, 1989.
Redondo et al, Science, 247:1225, 1990.
Reisman and Rotter, MoI Cell. Biol, 9:3571, 1989.
Resendez Jr. et al, MoI Cell. Biol, 8:4579, 1988.
Ripe et al, MoI Cell. Biol, 9:2224, 1989.
Rittling et al, Nuc. Acids Res., 17:1619, 1989.
RoIs and Teissie, Biophys. J., 75(3): 1415-1423, 1998.
Rosen et al, Cell, 41 :813, 1988.
Sakai et al, Genes and Dev., 2:1144, 1988.
Sambrook et al, In: Molecular cloning, Cold Spring Harbor Laboratory Press, Cold
Spring Harbor, NY, 2001. Satake et al, J. Virology, 62:970, 1988. Schaffher et al, J. MoI Biol, 201 :81, 1988. Searle et al, MoI Cell. Biol, 5:1480, 1985. Sharp and Marciniak, Cell, 59:229, 1989. Shaul and Ben-Levy, EMBO J., 6:1913, 1987. Sherman et al, MoI Cell. Biol, 9:50, 1989. Sleigh and Lockett, J. EMBO, 4:3831, 1985. Spalholz et al, Cell, 42:183, 1985. Spandau and Lee, J. Virology, 62:427, 1988. Spandidos and Wilkie, EMBO J., 2: 1193, 1983. Stephens and Hentschel, Biochem. J., 248:1, 1987. Strengall et al, J. Immunology, 170:5464-5469, 2003. Stuart et al, Nature, 317:828, 1985. Sullivan and Peterlin, MoI Cell. Biol, 7:3315, 1987. Swartzendruber and Lehman, J. Cell. Physiology, 85:179, 1975. Takebe et al., Mol. Cell. Biol., 8:466, 1988.
Tavernier et ah, Nature, 301 :634, 1983.
Taylor and Kingston, MoI. Cell. Biol, 10:165, 1990a.
Taylor and Kingston, MoI. Cell. Biol, 10:176, 1990b.
Taylor et al, J. Biol. Chem., 264:15160, 1989.
Thiesen et α/., J. Virology, 62:614, 1988.
Treisman, Cell, 42:889, 1985.
Tranche et al, MoI Biol. Med., 7:173, 1990.
Trudel and Constantini, Genes and Dev. 6:954, 1987.
Tsumaki et al, J. Biol. Chem., 273(36):22861-22864, 1998.
Tyndell et al, Nuc. Acids. Res., 9:6231, 1981.
Van Meirvenne et al, Cancer Gene Ther., 9(9):787-797, 2002. van Schooten et al, MoI Med Today, 3(6):254-260, 1997.
Van Tendeloo et al, Blood, 98(l):49-56, 2001.
Vannice and Levinson, J. Virology, 62:1305, 1988.
Vasseur et al, Proc Natl. Acad. Sci. U.S.A., 77:1068, 1980.
Wang and Calame, Cell, 47:241, 1986.
Weaver and Chizmadzhev, Bioenerg., 41 :135-160, 1996.
Weber et al, Cell, 36:983, 1984.
Weinberger et al MoI Cell. Biol, 8:988, 1984.
Weiss et al Proc. Am. Assoc. Cancer Res., 44(5493): 1094-1095, 2003.
Weiss et al Proc. Am. Assoc. Cancer Res., 44:1094-1095, 2003.
Wendtner, Leuk. Lymphoma, 45(5):897-904, 2004.
Wierda et al Blood, 96(9):2917-2924, 2000.
Winoto and Baltimore, Cell 59:649, 1989.
Wu et al, Biochem. Biophys. Res. Commun., 233(l):221-2266, 1997.
Yutzey et al MoI Cell. Biol, 9:1397, 1989.
Zhao-Emonet et al, Biochim. Biophys. Acta, 1442(2-3): 109-119, 1998.
Zhou et al, J. Biol. Chem., 270(21)12665-12669, 1995.
Lanier LL. Activating and inhibitory NK cell receptors. Adv Exp Med Biol. 1998;452:13-18.
Passweg JR, Tichelli A, Meyer-Monard S, et al. Purified donor NK- lymphocyte infusion to consolidate engraftment after haploidentical stem cell transplantation. Leukemia. 2004;18:1835-1838. McKenna DH, Jr., Sumstad D, Bostrom N, et al. Good manufacturing practices production of natural killer cells for immunotherapy: a six-year single-institution experience. Transfusion. 2007;47:520-528.
Chiorean EG, Miller JS. The biology of natural killer cells and implications for therapy of human disease. J Hematother Stem Cell Res. 2001;10:451-463.
Farag SS, Fehniger TA, Ruggeri L, Velardi A, Caligiuri MA. Natural killer cell receptors: new biology and insights into the graft- versus-leukemia effect. Blood. 2002;100:1935-1947.
Klingemann HG. Natural killer cell-based immunotherapeutic strategies. Cytotherapy. 2005;7: 16-22.
Miller JS, Soignier Y, Panoskaltsis-Mortari A, et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood. 2005;105:3051-3057.
Ruggeri L, Mancusi A, Burchielli E, Aversa F, Martelli MF, Velardi A. Natural killer cell alloreactivity in allogeneic hematopoietic transplantation. Curr Opin Oncol. 2007;19:142-147.
Imai C, Mihara K, Andreansky M, et al. Chimeric receptors with 4- IBB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia. 2004; 18:676-684.
Golzio M, RoIs MP, Teissie J. In vitro and in vivo electric field-mediated permeabilization, gene transfer, and expression. Methods. 2004;33:126-135.
Hui S, Li L. Electrically mediated delivery of molecules to cells: Human Press; 2000.
Xie TD, Sun L, Tsong TY. Study of mechanisms of electric field-induced DNA transfection. I. DNA entry by surface binding and diffusion through membrane pores. Biophys J. 1990;58: 13-19.
Zimmermann U. Electrical breakdown, electropermeabilization and electrofusion. Rev Physiol Biochem Pharmacol. 1986; 105: 176-256.
Li LH, Shivakumar R, Feller S, et al. Highly efficient, large volume flow electroporation. Technol Cancer Res Treat. 2002;l :341-350.
Trompeter HI, Weinhold S, Thiel C, Wernet P, Uhrberg M. Rapid and highly efficient gene transfer into natural killer cells by nucleofection. J Immunol Methods. 2003;274:245-256.
Li LH, Biagi E, Allen C, et al. Rapid and efficient nonviral gene delivery of CD 154 to primary chronic lymphocytic leukemia cells. Cancer Gene Ther. 2006; 13:215-224.
Li LH, McCarthy P, Hui SW. High-efficiency electrotransfection of human primary hematopoietic stem cells. Faseb J. 2001;15:586-588. Li LH, Sen A, Murphy SP, Jahreis GP, Fuji H, Hui SW. Apoptosis induced by DNA uptake limits transfection efficiency. Exp Cell Res. 1999;253:541-550.
Landi A, Babiuk LA, van Drunen Littel-van den Hurk S. High transfection efficiency, gene expression, and viability of monocyte-derived human dendritic cells after nonviral gene transfer. J Leukoc Biol. 2007.
Van De Parre TJ, Martinet W, Schrijvers DM, Herman AG, De Meyer GR. mRNA but not plasmid DNA is efficiently transfected in murine J774A.1 macrophages. Biochem Biophys Res Commun. 2005;327:356-360.
Rabinovich PM, Komarovskaya ME, Ye ZJ, et al. Synthetic messenger RNA as a tool for gene therapy. Hum Gene Ther. 2006;17:1027-1035.
Zhao Y, Zheng Z, Cohen CJ, et al. High-efficiency transfection of primary human and mouse T lymphocytes using RNA electroporation. MoI Ther. 2006; 13:151-159.
Manabe A, Coustan-Smith E, Kumagai M, et al. Interleukin-4 induces programmed cell death (apoptosis) in cases of high-risk acute lymphoblastic leukemia. Blood. 1994;83:1731-1737.
Ruggeri L, Capanni M, Urbani E, et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science. 2002;295:2097- 2100.
Leung W, Iyengar R, Turner V, et al. Determinants of antileukemia effects of allogeneic NK cells. J Immunol. 2004;172:644-650.
Caligiuri MA, Velardi A, Scheinberg DA, Borrello IM. Immunotherapeutic approaches for hematologic malignancies. Hematology Am Soc Hematol Educ Program. 2004:337-353.
Maasho K, Marusina A, Reynolds NM, Coligan JE, Borrego F. Efficient gene transfer into the human natural killer cell line, NKL, using the Amaxa nucleofection system. J Immunol Methods. 2004;284: 133-140.
Abbott BL. Recent advances in chronic lymphocytic leukemia. Cancer Invest. 2006;24:302-309.

Claims

1. A method for transiently modifying an unstimulated resting peripheral blood mononuclear cell (PBMC) to express a chimeric receptor on its surface comprising:
(a) isolating a resting PBMC;
(b) electroloading the PBMC with an mRNA encoding for a chimeric receptor, whereby the electroloaded PBMC transiently expresses the chimeric receptor on its surface.
2. The method of claim 1, wherein the PBMC is a peripheral blood lymphocyte (PBL).
3. The method of claim 2, wherein the PBL is a natural killer (NK) cell.
4. The method of claim 2, wherein the PBL is a T cell.
5. The method of claim 3, wherein isolating the NK cell comprises separation of CD56+ cells from blood.
6. The method of claim 3, wherein isolating the NK cell comprises depleting CD3+ cells from blood.
7. The method of claim 1, wherein the chimeric receptor is an anti-CD 19 chimeric receptor.
8. A method of treating cancer in a subject comprising:
(a) obtaining unstimulated resting peripheral blood mononuclear cells (PBMCs);
(b) electroloading the PBMCs with an mRNA encoding for a chimeric receptor, whereby the electrotransfected PBMCs transiently express the chimeric receptor on their surface; and
(c) administering the trans fected PBMCs to the subject to treat cancer in the subject.
9. The method of claim 8, wherein the PMBCs are peripheral blood lymphocytes (PBLs).
10. The method of claim 9, wherein the PBLs are natural killer (NK) cells.
11. The method of claim 8, wherein the subject is a human.
12. The method of claim 8, wherein the PMBCs are autologous to the subject.
13. The method of claim 8, wherein the PMBCs are allogenic to the subject.
14. The method of claim 8, wherein obtaining isolated PMBCs comprises collecting peripheral blood from a donor and separating the PMBCs in the blood from non-PMBCs in the blood.
15. The method of claim 14, wherein the donor is also the subject being treated.
16. The method of claim 14, wherein the time from when the peripheral blood is collected from the donor to the time when the transfected PMBCs are administered to the subject is less than 48 hours.
17. The method of claim 16, wherein the time is less that 12 hours.
18. The method of claim 8, wherein the cancer is a leukemia.
19. The method of claim 8, wherein the chimeric receptor is an anti-CD 19 chimeric receptor.
20. The method of claim 8, wherein the transfected PMBCs are administered by intravenous injection.
21. The method of claim 8, wherein the transfected PMBCs are administered by intratumoral injection.
22. A composition comprising:
(a) an electrotransfected unstimulated resting peripheral blood lymphocyte
(PBL) transiently expressing an mRNA coding for a chimeric receptor, whereby the chimeric receptor is expressed on the surface of the electrotransfected PBL; and
(b) a pharmaceutically acceptable carrier.
23. The composition of claim 22, wherein the PBL is a natural killer (NK) cell.
24. The composition of claim 22, wherein the chimeric receptor is an anti-CD 19 chimeric receptor.
PCT/US2009/040040 2008-04-09 2009-04-09 Engineering and delivery of therapeutic compositions of freshly isolated cells WO2009126789A2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
DK09731422.3T DK2279253T3 (en) 2008-04-09 2009-04-09 Construction and application of therapeutic compositions of freshly isolated cells
JP2011504171A JP5779090B2 (en) 2008-04-09 2009-04-09 Manipulation and delivery of therapeutic compositions of newly isolated cells
EP09731422.3A EP2279253B1 (en) 2008-04-09 2009-04-09 Engineering and delivery of therapeutic compositions of freshly isolated cells

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US4365308P 2008-04-09 2008-04-09
US61/043,653 2008-04-09

Publications (2)

Publication Number Publication Date
WO2009126789A2 true WO2009126789A2 (en) 2009-10-15
WO2009126789A3 WO2009126789A3 (en) 2010-01-28

Family

ID=41162602

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/040040 WO2009126789A2 (en) 2008-04-09 2009-04-09 Engineering and delivery of therapeutic compositions of freshly isolated cells

Country Status (5)

Country Link
US (8) US8450112B2 (en)
EP (1) EP2279253B1 (en)
JP (4) JP5779090B2 (en)
DK (1) DK2279253T3 (en)
WO (1) WO2009126789A2 (en)

Families Citing this family (67)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9249423B2 (en) 2007-02-02 2016-02-02 Yale University Method of de-differentiating and re-differentiating somatic cells using RNA
US8859229B2 (en) * 2007-02-02 2014-10-14 Yale University Transient transfection with RNA
US10155038B2 (en) 2007-02-02 2018-12-18 Yale University Cells prepared by transient transfection and methods of use thereof
DK2279253T3 (en) * 2008-04-09 2017-02-13 Maxcyte Inc Construction and application of therapeutic compositions of freshly isolated cells
AP2013006918A0 (en) * 2010-12-09 2013-06-30 Univ Pennsylvania Use of chimeric antigen receptormodified T-cells to treat cancer
JP5572863B2 (en) * 2011-06-24 2014-08-20 国立大学法人九州大学 Method for amplifying NK cells
JP5989016B2 (en) * 2011-06-24 2016-09-07 国立大学法人九州大学 Method for amplifying NK cells
CA2848410A1 (en) 2011-09-16 2013-03-21 The Trustees Of The University Of Pennsylvania Rna engineered t cells for the treatment of cancer
WO2013063419A2 (en) * 2011-10-28 2013-05-02 The Trustees Of The University Of Pennsylvania A fully human, anti-mesothelin specific chimeric immune receptor for redirected mesothelin-expressing cell targeting
CA3209571A1 (en) 2012-03-23 2013-09-26 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Anti-mesothelin chimeric antigen receptors
US10144770B2 (en) 2013-10-17 2018-12-04 National University Of Singapore Chimeric receptors and uses thereof in immune therapy
SG11201608395PA (en) 2014-04-10 2016-11-29 Seattle Children S Hospital Dba Seattle Children S Res Inst Defined composition gene modified t-cell products
WO2016095934A2 (en) * 2014-12-14 2016-06-23 El Abd Hisham Mohamed Magdy A novel genetic device to engineer cell behavior
CN107429254B (en) 2015-01-30 2021-10-15 加利福尼亚大学董事会 Protein delivery in primary hematopoietic cells
WO2017004252A1 (en) 2015-06-30 2017-01-05 The Arizona Board Of Regents On Behalf Of The University Of Arizona Redirected cells with mhc chimeric receptors and methods of use in immunotherapy
KR20180028533A (en) 2015-07-28 2018-03-16 더 트러스티스 오브 더 유니버시티 오브 펜실바니아 Modified Monocytes / Macrophages and Their Uses Expressing Chimeric Antigen Receptors
US11458167B2 (en) 2015-08-07 2022-10-04 Seattle Children's Hospital Bispecific CAR T-cells for solid tumor targeting
EP3708587A1 (en) 2015-11-27 2020-09-16 Cartherics Pty. Ltd. Genetically modified cells and uses thereof
US11293021B1 (en) 2016-06-23 2022-04-05 Inscripta, Inc. Automated cell processing methods, modules, instruments, and systems
CA3045665A1 (en) 2016-12-12 2018-06-21 Seattle Children's Hospital (dba Seattle Children's Research Institute) Chimeric transcription factor variants with augmented sensitivity to drug ligand induction of transgene expression in mammalian cells
CN110612119A (en) 2017-02-07 2019-12-24 西雅图儿童医院(Dba西雅图儿童研究所) Phospholipid ether (PLE) CAR T cell tumor targeting (CTCT) agents
US11850262B2 (en) 2017-02-28 2023-12-26 Purdue Research Foundation Compositions and methods for CAR T cell therapy
AU2018256436A1 (en) 2017-04-19 2019-11-07 Board Of Regents, The University Of Texas System Immune cells expressing engineered antigen receptors
US9982279B1 (en) 2017-06-23 2018-05-29 Inscripta, Inc. Nucleic acid-guided nucleases
US10011849B1 (en) 2017-06-23 2018-07-03 Inscripta, Inc. Nucleic acid-guided nucleases
ES2913457T3 (en) 2017-06-30 2022-06-02 Inscripta Inc Automated Cell Processing Methods, Modules, Instruments and Systems
CN111094330A (en) * 2017-07-03 2020-05-01 转矩医疗股份有限公司 Polynucleotides encoding immunostimulatory fusion molecules and uses thereof
US10738327B2 (en) 2017-08-28 2020-08-11 Inscripta, Inc. Electroporation cuvettes for automation
JP6985508B2 (en) 2017-09-30 2021-12-22 インスクリプタ, インコーポレイテッド Flow-through electroporation equipment organization
WO2019090355A1 (en) * 2017-11-06 2019-05-09 Children's National Medical Center Cells expressing antibodies and methods of treatment using the same
KR20200118010A (en) * 2018-01-05 2020-10-14 맥스시티 인코포레이티드 Chronic CAR treatment of cancer
AU2019209428A1 (en) 2018-01-22 2020-07-30 Endocyte, Inc. Methods of use for CAR T cells
EP3749753A1 (en) * 2018-02-09 2020-12-16 Immatics US, Inc. Methods for manufacturing t cells
AU2019241967A1 (en) 2018-03-29 2020-11-19 Inscripta, Inc. Automated control of cell growth rates for induction and transformation
WO2019200004A1 (en) 2018-04-13 2019-10-17 Inscripta, Inc. Automated cell processing instruments comprising reagent cartridges
US10858761B2 (en) 2018-04-24 2020-12-08 Inscripta, Inc. Nucleic acid-guided editing of exogenous polynucleotides in heterologous cells
US10501738B2 (en) 2018-04-24 2019-12-10 Inscripta, Inc. Automated instrumentation for production of peptide libraries
US10557216B2 (en) 2018-04-24 2020-02-11 Inscripta, Inc. Automated instrumentation for production of T-cell receptor peptide libraries
EP4070802A1 (en) 2018-06-30 2022-10-12 Inscripta, Inc. Instruments, modules, and methods for improved detection of edited sequences in live cells
US11142740B2 (en) 2018-08-14 2021-10-12 Inscripta, Inc. Detection of nuclease edited sequences in automated modules and instruments
US10532324B1 (en) 2018-08-14 2020-01-14 Inscripta, Inc. Instruments, modules, and methods for improved detection of edited sequences in live cells
AU2019336229A1 (en) * 2018-09-07 2021-03-18 Sotio, LLC Chimeric receptor polypeptides in combination with trans metabolism molecules modulating intracellular lactate concentrations and therapeutic uses thereof
CN113227368B (en) 2018-10-22 2023-07-07 因思科瑞普特公司 Engineered enzymes
US11214781B2 (en) 2018-10-22 2022-01-04 Inscripta, Inc. Engineered enzyme
CN111349615A (en) * 2018-12-24 2020-06-30 上海细胞治疗集团有限公司 Method for preparing cell over expressing exogenous gene
CN113631713A (en) 2019-03-25 2021-11-09 因思科瑞普特公司 Simultaneous multiplex genome editing in yeast
US11001831B2 (en) 2019-03-25 2021-05-11 Inscripta, Inc. Simultaneous multiplex genome editing in yeast
CN113939593A (en) 2019-06-06 2022-01-14 因思科瑞普特公司 Treatment for recursive nucleic acid-guided cell editing
CA3139124C (en) 2019-06-21 2023-01-31 Inscripta, Inc. Genome-wide rationally-designed mutations leading to enhanced lysine production in e. coli
US10927385B2 (en) 2019-06-25 2021-02-23 Inscripta, Inc. Increased nucleic-acid guided cell editing in yeast
WO2021102059A1 (en) 2019-11-19 2021-05-27 Inscripta, Inc. Methods for increasing observed editing in bacteria
WO2021118626A1 (en) 2019-12-10 2021-06-17 Inscripta, Inc. Novel mad nucleases
US10704033B1 (en) 2019-12-13 2020-07-07 Inscripta, Inc. Nucleic acid-guided nucleases
EP4069851A4 (en) 2019-12-18 2023-11-22 Inscripta, Inc. Cascade/dcas3 complementation assays for in vivo detection of nucleic acid-guided nuclease edited cells
US10689669B1 (en) 2020-01-11 2020-06-23 Inscripta, Inc. Automated multi-module cell processing methods, instruments, and systems
CN115135370A (en) 2020-01-27 2022-09-30 因思科瑞普特公司 Electroporation module and instrument
US20210332388A1 (en) 2020-04-24 2021-10-28 Inscripta, Inc. Compositions, methods, modules and instruments for automated nucleic acid-guided nuclease editing in mammalian cells
US11787841B2 (en) 2020-05-19 2023-10-17 Inscripta, Inc. Rationally-designed mutations to the thrA gene for enhanced lysine production in E. coli
KR20230042691A (en) 2020-06-04 2023-03-29 카리스마 테라퓨틱스 인코포레이티드 Novel constructs for chimeric antigen receptors
WO2022060749A1 (en) 2020-09-15 2022-03-24 Inscripta, Inc. Crispr editing to embed nucleic acid landing pads into genomes of live cells
US11512297B2 (en) 2020-11-09 2022-11-29 Inscripta, Inc. Affinity tag for recombination protein recruitment
AU2021415461A1 (en) 2021-01-04 2023-08-17 Inscripta, Inc. Mad nucleases
EP4274890A1 (en) 2021-01-07 2023-11-15 Inscripta, Inc. Mad nucleases
US11884924B2 (en) 2021-02-16 2024-01-30 Inscripta, Inc. Dual strand nucleic acid-guided nickase editing
EP4279085A1 (en) 2022-05-20 2023-11-22 Mnemo Therapeutics Compositions and methods for treating a refractory or relapsed cancer or a chronic infectious disease
WO2024059618A2 (en) 2022-09-13 2024-03-21 Arsenal Biosciences, Inc. Immune cells having co-expressed tgfbr shrnas
WO2024059824A2 (en) 2022-09-16 2024-03-21 Arsenal Biosciences, Inc. Immune cells with combination gene perturbations

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US5612207A (en) 1993-03-23 1997-03-18 Cbr Laboratories, Inc. Method and apparatus for encapsulation of biologically-active substances in cells
US5720921A (en) 1995-03-10 1998-02-24 Entremed, Inc. Flow electroporation chamber and method
US5925565A (en) 1994-07-05 1999-07-20 Institut National De La Sante Et De La Recherche Medicale Internal ribosome entry site, vector containing it and therapeutic use
US5928906A (en) 1996-05-09 1999-07-27 Sequenom, Inc. Process for direct sequencing during template amplification
US5935819A (en) 1992-08-27 1999-08-10 Eichner; Wolfram Process for producing a pharmaceutical preparation of PDGF-AB
US6074605A (en) 1995-03-10 2000-06-13 Entremed, Inc. Flow electroporation chamber and method
US6090617A (en) 1996-12-05 2000-07-18 Entremed, Inc. Flow electroporation chamber with electrodes having a crystalline metal nitride coating
WO2003018751A2 (en) 2001-08-22 2003-03-06 Maxcyte, Inc. Apparatus and method for electroporation of biological samples
US20040038886A1 (en) 2000-10-16 2004-02-26 Finney Helene Margaret Chimeric cytoplasmic signalling molecules derived from cd137
WO2004031353A2 (en) 2002-09-30 2004-04-15 Maxcyte, Inc. Apparatus and method for streaming electroporation
US6773669B1 (en) 1995-03-10 2004-08-10 Maxcyte, Inc. Flow electroporation chamber and method
US8027202B2 (en) 2008-04-11 2011-09-27 Hynix Semiconductor Inc. Method of programming a flash memory device using self boosting

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6103521A (en) 1995-02-06 2000-08-15 Cell Genesys, Inc. Multispecific chimeric receptors
US7446179B2 (en) * 2000-11-07 2008-11-04 City Of Hope CD19-specific chimeric T cell receptor
US7029916B2 (en) 2001-02-21 2006-04-18 Maxcyte, Inc. Apparatus and method for flow electroporation of biological samples
US7723111B2 (en) 2001-03-09 2010-05-25 The United States Of America As Represented By The Department Of Health And Human Services Activated dual specificity lymphocytes and their methods of use
EP1597390A4 (en) 2003-02-18 2006-12-27 Maxcyte Inc Loading of cells with antigens by electroporation
WO2006063301A1 (en) * 2004-12-10 2006-06-15 Maxcyte, Inc. Genetically modified tumor cells as cancer vaccines
EP1795599A1 (en) * 2005-12-09 2007-06-13 Schuler, Gerold, Prof. Dr. Methods for generating antigen-specific effector T cells
US8859229B2 (en) * 2007-02-02 2014-10-14 Yale University Transient transfection with RNA
WO2008121420A1 (en) * 2007-03-30 2008-10-09 Memorial Sloan-Kettering Cancer Center Constitutive expression of costimulatory ligands on adoptively transferred t lymphocytes
DK2279253T3 (en) * 2008-04-09 2017-02-13 Maxcyte Inc Construction and application of therapeutic compositions of freshly isolated cells
WO2012050374A2 (en) 2010-10-13 2012-04-19 Innocell, Inc. Immunotherapy for solid tumors
AP2013006918A0 (en) 2010-12-09 2013-06-30 Univ Pennsylvania Use of chimeric antigen receptormodified T-cells to treat cancer
EP3974520A1 (en) 2013-05-14 2022-03-30 Board of Regents, The University of Texas System Human application of engineered chimeric antigen receptor (car) t-cells
EP3685842B1 (en) 2014-06-06 2022-08-31 Memorial Sloan-Kettering Cancer Center Mesothelin-targeted chimeric antigen receptors and uses thereof
CN116426490A (en) 2016-03-19 2023-07-14 埃克苏马生物技术公司 Methods and compositions for modulation of lymphocyte transduction and expansion thereof
KR20200118010A (en) 2018-01-05 2020-10-14 맥스시티 인코포레이티드 Chronic CAR treatment of cancer

Patent Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4683202B1 (en) 1985-03-28 1990-11-27 Cetus Corp
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US5935819A (en) 1992-08-27 1999-08-10 Eichner; Wolfram Process for producing a pharmaceutical preparation of PDGF-AB
US5612207A (en) 1993-03-23 1997-03-18 Cbr Laboratories, Inc. Method and apparatus for encapsulation of biologically-active substances in cells
US5925565A (en) 1994-07-05 1999-07-20 Institut National De La Sante Et De La Recherche Medicale Internal ribosome entry site, vector containing it and therapeutic use
US6773669B1 (en) 1995-03-10 2004-08-10 Maxcyte, Inc. Flow electroporation chamber and method
US5720921A (en) 1995-03-10 1998-02-24 Entremed, Inc. Flow electroporation chamber and method
US6074605A (en) 1995-03-10 2000-06-13 Entremed, Inc. Flow electroporation chamber and method
US5928906A (en) 1996-05-09 1999-07-27 Sequenom, Inc. Process for direct sequencing during template amplification
US6090617A (en) 1996-12-05 2000-07-18 Entremed, Inc. Flow electroporation chamber with electrodes having a crystalline metal nitride coating
US6617154B1 (en) 1996-12-05 2003-09-09 Maxcyte, Inc. Electroporation chamber including an electrode having a continuous, crystalline metal nitride coating
US6485961B1 (en) 1996-12-05 2002-11-26 Maxcyte, Inc. Electrodes having a continuous, crystalline metal nitride coating and method of use
US20040038886A1 (en) 2000-10-16 2004-02-26 Finney Helene Margaret Chimeric cytoplasmic signalling molecules derived from cd137
WO2003018751A2 (en) 2001-08-22 2003-03-06 Maxcyte, Inc. Apparatus and method for electroporation of biological samples
WO2004031353A2 (en) 2002-09-30 2004-04-15 Maxcyte, Inc. Apparatus and method for streaming electroporation
US8027202B2 (en) 2008-04-11 2011-09-27 Hynix Semiconductor Inc. Method of programming a flash memory device using self boosting

Non-Patent Citations (241)

* Cited by examiner, † Cited by third party
Title
ABBOTT BL: "Recent advances in chronic lymphocytic leukemia", CANCER INVEST., vol. 24, 2006, pages 302 - 309
ALMENDRO ET AL., J. IMMUNOL., vol. 157, no. 12, 1996, pages 5411 - 5421
ANGEL ET AL., CELL, vol. 49, 1987, pages 729
ANGEL ET AL., MOL. CELL. BIOL., vol. 7, 1987, pages 2256
ATCHISON; PERRY, CELL, vol. 46, 1986, pages 253
ATCHISON; PERRY, CELL, vol. 48, 1987, pages 121
AUSUBEL ET AL.: "Current Protocols in Molecular Biology", 1996, JOHN, WILEY & SONS, INC
BANERJI ET AL., CELL, vol. 27, 1981, pages 299 - 308
BANERJI ET AL., CELL, vol. 33, no. 3, 1983, pages 729 - 740
BERKHOUT ET AL., CELL, vol. 59, 1989, pages 273 - 282
BIAGI ET AL., HUM. GEN THER., vol. 14, no. 6, 2003, pages 545 - 559
BIOENERG, vol. 45, 1998, pages 3 - 16
BLANAR ET AL., EMBO J., vol. 8, 1989, pages 1139
BODINE; LEY, EMBO J., vol. 6, 1987, pages 2997
BOISSEL LAURENT;: "Transfection with mRNA for CD19 specific chimeric antigen receptor restores natural killer cell mediated killing of CLL cells", BLOOD, vol. 110, no. 11, 16 November 2007 (2007-11-16), pages 915A
BOSHART ET AL., CELL, vol. 41, 1985, pages 521
BOSZE ET AL., EMBO J., vol. 5, no. 7, 1986, pages 1615 - 1623
BRADDOCK ET AL., CELL, vol. 58, 1989, pages 269
BULLA; SIDDIQUI, J. VIROL., vol. 62, 1986, pages 1437
CALIFANO ET AL., CANCER RES., vol. 56, no. 11, 1996, pages 2488 - 2492
CAMPBELL; VILLARREAL, MOL. CELL. BIOL., vol. 8, 1988, pages 1993
CAMPERE; TILGHMAN, GENES AND DEV., vol. 3, 1989, pages 537
CAMPO ET AL., NATURE, vol. 303, 1983, pages 77
CARBONELLI ET AL., FEMS MICROBIOL. LETT., vol. 177, no. I, 1999, pages 75 - 82
CELANDER ET AL., J. VIROLOGY,, vol. 62, 1988, pages 1314
CELANDER; HASELTINE, J. VIROLOGY, vol. 61, 1987, pages 269
CHANDLER ET AL., CELL, vol. 33, 1983, pages 489
CHANDLER ET AL., PROC. NATL. ACAD. SEI. USA, vol. 94, no. 8, 1997, pages 3596 - 601
CHANG ET AL., MOL. CELL. BIOL., vol. 9, 1989, pages 2153
CHATTERJEE ET AL., PROC. NATL. ACAD. SEI. USA, vol. 86, 1989, pages 9114
CHEN ET AL., ONCOGENE, vol. 8, 1993, pages 2159 - 66
CHIOREAN EG; MILLER JS: "The biology of natural killer cells and implications for therapy of human disease", J HEMATOTHER STEM CELL RES, vol. 10, 2001, pages 451 - 463
CHOI ET AL., CELL, vol. 53, 1988, pages 519
COCEA, BIOTECHNIQUES, vol. 23, no. 5, 1997, pages 814 - 816
COHEN ET AL., J. CELL. PHYSIOL., vol. 5, 1987, pages 75
CONNOR ET AL., INT. J. GYNECOL. CANCER, vol. 3, no. 2, 1993, pages 103 - 109
CONNOR ET AL., J. EXP. MED., vol. 177, 1993, pages 1127 - 1134
COSTA ET AL., MOL. CELL. BIOL., vol. 8, 1988, pages 81
CRIPE ET AL., EMBO J., vol. 6, 1987, pages 3745
CULOTTA; HAMER, MOL. CELL. BIOL., vol. 9, 1989, pages 1376
DANDOLO ET AL., J. VIROLOGY, vol. 47, 1983, pages 55 - 64
DATABASE BIOSIS accession no. REV200800218382
DE JAGER ET AL., SEMIN. NUCL. MED., vol. 23, no. 2, 1993, pages 165 - 179
DE VILLIERS ET AL., NATURE, vol. 312, no. 5991, 1984, pages 242 - 246
DESCHAMPS ET AL., SCIENCE, vol. 230, 1985, pages 1174 - 1177
DILLOO ET AL., BLOOD, vol. 90, no. 5, 1997, pages 1927 - 1933
DOOLITTLE; BEN-ZEEV, METHODS MOL BIOL, vol. 109, 1999, pages 215 - 237
DRANOFF ET AL., PROC. NATL. ACAD. SEI. USA, vol. 90, 1993, pages 3539 - 3543
EDBROOKE ET AL., MOL. CELL. BIOL, vol. 9, 1989, pages 1908
EDLUND ET AL., SCIENCE, vol. 230, 1985, pages 912 - 916
FARAG S S,ET AL: "Natural killer cell receptors: New biology and insights into the graft-versus-leukemia effect", BLOOD, AMERICAN SOCIETY OF HEMATOLOGY, & 55TH ANNUAL MEETING OF THE AMERICAN-SOCIETY-OF-HEMATOLOGY; NEW ORLEANS, LA, USA; DECEMBER 07 -10, 2013, vol. 100, no. 6, 15 September 2002 (2002-09-15), & 55TH ANNUAL MEETING OF THE AMERICAN-SOCIETY-OF-HEMATOLOGY; NEW ORLEANS, LA, USA; DECEMBER 07 -10, 2013, pages 1935 - 1947, XP002308880, ISSN: 0006-4971, DOI: 10.1182/blood-2002-02-0350
FEARON; VOGELSTEIN, CELL, vol. 61, no. 5, 1990, pages 759 - 767
FENG; HOLLAND, NATURE, vol. 334, 1988, pages 6178
FIRAK; SUBRAMANIAN, MOL. CELL. BIOL., vol. 6, 1986, pages 3667
FOECKING; HOFSTETTER, GENE, vol. 45, no. 1, 1986, pages 101 - 105
FUJITA ET AL., CELL, vol. 49, 1987, pages 357
GABRIEL; TEISSIE, BIOPHYS. J., vol. 76, no. 4, 1999, pages 2158 - 2165
GANSBACHER ET AL., CANCER RES., vol. 50, no. 24, 1990, pages 7820 - 7825
GANSBACHER ET AL., J. EXP. MED., vol. 172, 1990, pages 1217 - 1224
GILBOA E., J CLIN INVEST., vol. 7, no. 5, 11 May 2007 (2007-05-11), pages 1195 - 203
GILLES ET AL., CELL, vol. 33, 1983, pages 717
GLOSS ET AL., EMBO J., vol. 6, 1987, pages 3735
GODBOUT ET AL., MOL. CELL. BIOL., vol. 8, 1988, pages 1169
GOLZIO, M. ROLS, M.P. TEISSIE, J.: "In vitro and in vivo electric field-mediated permeabilization, gene transfer, and expression", METHODS, ACADEMIC PRESS, NL, vol. 33, no. 2, 1 June 2004 (2004-06-01), NL, pages 126 - 135, XP004505454, ISSN: 1046-2023, DOI: 10.1016/j.ymeth.2003.11.003
GOODBOUM ET AL., CELL, vol. 45, 1986, pages 601
GOODBOUM; MANIATIS, PROC. NATL. ACAD. SEI. USA, vol. 85, 1988, pages 1447
GREENE ET AL., IMMUNOLOGY TODAY, vol. 10, 1989, pages 272
GROSSCHEDL; BALTIMORE, CELL, vol. 41, 1985, pages 885
GULBIS; GALAND, HUM. PATHOL., vol. 24, no. 12, 1993, pages 1271 - 1285
HARDING, EUR. J. IMMUNOL., vol. 22, no. 7, 1992, pages 1865 - 1869
HASLINGER; KARIN, PROC. NATL. ACAD. SEI. USA, vol. 82, 1985, pages 8572
HAUBER; CULLEN, J. VIROLOGY, vol. 62, 1988, pages 673
HEN ET AL., NATURE, vol. 321, 1986, pages 249
HENSEL ET AL., LYMPHOKINE RES., vol. 8, 1989, pages 347
HERR; CLARKE, CELL, vol. 45, 1986, pages 461
HIBINO ET AL., BIOPHYS. J., vol. 64, no. 6, 1993, pages 1789 - 1800
HIROCHIKA ET AL., J. VIROL., vol. 61, 1987, pages 2599
HIRSCH ET AL., MOL. CELL. BIOL., vol. 10, 1990, pages 1959
HOLBROOK ET AL., VIROLOGY, vol. 157, 1987, pages 211
HONG C; PARK SH, CRIT REV IMMUNOL., vol. 27, no. 6, 2007, pages 511 - 25
HORLICK; BENFIELD, MOL. CELL. BIOL., vol. 9, 1989, pages 2396
HUANG ET AL., CELL, vol. 27, 1981, pages 245
HUG ET AL., MOL. CELL. BIOL., vol. 8, 1988, pages 3065
HUI S; FI F: "Electrically mediated delivery of molecules to cells", 2000, HUMAN PRESS
HWANG ET AL., MOL. CELL. BIOL., vol. 10, 1990, pages 585
IMAGAWA ET AL., CELL, vol. 51, 1987, pages 251
IMAI C, ET AL: "Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia.", LEUKEMIA, vol. 18, no. 4, 1 April 2004 (2004-04-01), pages 676 - 684, XP002579934, ISSN: 0887-6924
IMBRA; KARIN, NATURE, vol. 323, 1986, pages 555
IMLER ET AL., MOL. CELL. BIOL., vol. 7, 1987, pages 2558
IMPERIALE; NEVINS, MOL. CELL. BIOI, vol. 4, 1984, pages 875
JAIN ET AL., ANN. SURG. ONCOLOGY, vol. 10, 2003, pages 810 - 820
JAKOBOVITS ET AL., MOL. CELL. BIOL., vol. 8, 1988, pages 2555
JAMEEL; SIDDIQUI, MOL. CELL. BIOL., vol. 6, 1986, pages 710
JAYNES ET AL., MOL. CELL. BIOL., vol. 8, 1988, pages 62
JOHNSON ET AL., MOL. CELL. BIOL., vol. 9, 1989, pages 3393
KADESCH; BERG, MOL. CELL. BIOL., vol. 6, 1986, pages 2593
KAKORIN ET AL., BIOPHYS. CHEM., vol. 58, no. 1-2, 1996, pages 109 - 116
KARIN ET AL., MOL. CELL. BIOL., vol. 7, 1987, pages 606
KARP ET AL., J. IMMUNOL., vol. 150, no. 3, 1993, pages 896 - 908
KASAIAN ET AL., IMMUNITY, vol. 16, 2002, pages 559 - 569
KATINKA, CELL, vol. 20, 1980, pages 393
KAWAMOTO ET AL., MOL. CELL. BIOL., vol. 8, 1988, pages 267
KILEDJIAN ET AL., MOL. CELL. BIOL.,, vol. 8, 1988, pages 145
KIM ET AL., J. VIROL., vol. 76, no. 4, 2002, pages 1892 - 1903
KIMURA ET AL., ARCH. OTOLARYNGOL HEAD NECK SURG., vol. 129, 2003, pages 1181 - 1185
KISHIDA ET AL., GENE THERAPY, vol. 8, 2001, pages 1234 - 1240
KISHIDA ET AL., MOL. THER., vol. 8, no. 4, 2003, pages 552 - 558
KLAMUT ET AL., MOL. CELL. BIOL., vol. 10, 1990, pages 193
KLINGEMANN H-G: "Natural killer cell-based immunotherapeutic strategies", CYTOTHERAPY, ISIS MEDICAL MEDIA, OXFORD,, GB, vol. 7, no. 1, 1 January 2005 (2005-01-01), GB, pages 16 - 22, XP009089974, ISSN: 1465-3249
KNUTSON; YEE, ANAL. BIOCHEM., vol. 164, no. 1, 1987, pages 44 - 52
KOCH ET AL., MOL. CELL. BIOL., vol. 9, 1989, pages 303
KOTNIK ET AL., BIOENERG, vol. 43, pages 281 - 291
KRAUS ET AL., . FEBSLETT., vol. 428, no. 3, 1998, pages 165 - 170
KRIEGLER ET AL., CELL, vol. 38, 1984, pages 483
KRIEGLER ET AL., CELL, vol. 53, 1988, pages 45
KRIEGLER; BOTCHAN, MOL. CELL. BIOL., vol. 3, 1983, pages 325
KRIEGLER; BOTCHAN: "Eukaryotic Viral Vectors", 1982
KÜHL ET AL., CELL, vol. 50, 1987, pages 1057
KUNZ ET AL., NUCL. ACIDS RES., vol. 17, 1989, pages 1121
LANDI A; BABIUK LA; VAN DRUNEN LITTEL-VAN DEN HURK S: "High transfection efficiency, gene expression, and viability of monocyte-derived human dendritic cells after nonviral gene transfer", J LEUKOC BIOL., 2007
LANIER LL: "Activating and inhibitory NK cell receptors", ADV EXP MED BIOL, vol. 452, 1998, pages 13 - 18
LAREYRE ET AL., J. BIOL. CHEM., vol. 274, no. 12, 1999, pages 8282 - 8290
LARSEN ET AL., PROC NATL. ACAD. SEI. USA., vol. 83, 1986, pages 8283
LASPIA ET AL., CELL, vol. 59, 1989, pages 283
LATIMER ET AL., MOL. CELL. BIOL., vol. 10, 1990, pages 760
LE BLANC K; RINGDEN O, INTERN MED., vol. 2, no. 5, 26 November 2007 (2007-11-26), pages 509 - 25
LEE ET AL., DNA CELL BIOL., vol. 16, no. 11, 1997, pages 1267 - 1275
LEE ET AL., NATURE, vol. 294, 1981, pages 228
LEE ET AL., NUCLEIC ACIDS RES., vol. 12, 1984, pages 4191 - 206
LEUNG W, ET AL: "Determinants of antileukemia effects of allogeneic NK cells.", THE JOURNAL OF IMMUNOLOGY, THE AMERICAN ASSOCIATION OF IMMUNOLOGISTS, INC., US, vol. 172, no. 1, 1 January 2004 (2004-01-01), US, pages 644 - 650, XP002382435, ISSN: 0022-1767
LEVENSON ET AL., HUM. GENE THER, vol. 9, no. 8, 1998, pages 1233 - 1236
LEVINSON ET AL., NATURE, vol. 295, 1982, pages 79
LI ET AL., AM. SOC. GENE THERAPY, 2002
LI ET AL., J. LEUKOC. BIOL.,, vol. 56, no. 5, 1994, pages 616 - 624
LI LH; BIAGI E; ALLEN C ET AL.: "Rapid and efficient nonviral gene delivery of CD 154 to primary chronic lymphocytic leukemia cells", CANCER GENE THER., vol. 13, 2006, pages 215 - 224
LI LH; MCCARTHY P; HUI SW: "High-efficiency electrotransfection of human primary hematopoietic stem cells", FASEB J., vol. 15, 2001, pages 586 - 588
LI LH; SEN A; MURPHY SP; JAHREIS GP; FUJI H; HUI SW.: "Apoptosis induced by DNA uptake limits transfection efficiency", EXP CELL RES, vol. 253, 1999, pages 541 - 550
LI LIN-HONG; SHIVAKUMAR RAMA; FELLER STEPHANIE; ALLEN CORNELL; WEISS JONATHAN M; DZEKUNOV SERGEY; SINGH VIN; HOLADAY JOHN; FRATANT: "HIGHLY EFFICIENT, LARGE VOLUME FLOW ELECTROPORATION", TECHNOLOGY IN CANCER RESEARCH AND TREATMENT, ADENINE PRESS, SCHENECTADY, NY, US, vol. 1, no. 5, 1 October 2002 (2002-10-01), US, pages 341 - 350, XP009083609, ISSN: 1533-0346
LIN ET AL., MOL. CELL. BIOL., vol. 10, 1990, pages 850
LURIA ET AL., EMBO J., vol. 6, 1987, pages 3307
LUSKY ET AL., MOL. CELL. BIOL., vol. 3, 1983, pages 1108
LUSKY; BOTCHAN, PROC. NATL. ACAD. SEI. USA, vol. 83, 1986, pages 3609
M. A. CALIGIURI, ANDREA VELARDI, DAVID A SCHEINBERG, IVAN M BORRELLO: "Immunotherapeutic Approaches for Hematologic Malignancies", HEMATOLOGY, vol. 2004, no. 1, 1 January 2004 (2004-01-01), pages 337 - 353, XP055186287, ISSN: 15204391, DOI: 10.1182/asheducation-2004.1.337
MA ET AL., J. IMMUNOL., vol. 171, no. 2, 2003, pages 608 - 615
MAASHO, K. MARUSINA, A. REYNOLDS, N.M. COLIGAN, J.E. BORREGO, F.: "Efficient gene transfer into the human natural killer cell line, NKL, using the Amaxa nucleofection system(TM)", JOURNAL OF IMMUNOLOGICAL METHODS., ELSEVIER SCIENCE PUBLISHERS B.V.,AMSTERDAM., NL, vol. 284, no. 1-2, 1 January 2004 (2004-01-01), NL, pages 133 - 140, XP004485592, ISSN: 0022-1759, DOI: 10.1016/j.jim.2003.10.010
MACEJAK; SAMOW, NATURE, vol. 353, 1991, pages 90 - 94
MAJORS; VARMUS, PROC. NATL. ACAD. SCI. USA, vol. 80, 1983, pages 5866
MANABE A; COUSTAN-SMITH E; KUMAGAI M ET AL.: "Interleukin-4 induces programmed cell death (apoptosis) in cases of high-risk acute lymphoblastic leukemia", BLOOD, vol. 83, 1994, pages 1731 - 1737
MARTINO A; POCCIA F, CURR MOL MED., 7 November 2007 (2007-11-07), pages 658 - 73
MCKENNA DH, JR.; SUMSTAD D; BOSTROM N ET AL.: "Good manufacturing practices production of natural killer cells for immunotherapy: a six-year single-institution experience", TRANSFUSION, vol. 47, 2007, pages 520 - 528
MCNEALL ET AL., GENE, vol. 76, 1989, pages 81
MELLMAN; STEINMAN, CELL, vol. 106, no. 3, 2001, pages 255 - 258
MIKSICEK ET AL., CELL, vol. 46, 1986, pages 203
MILLER JEFFREY S ET AL: "Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer", BLOOD, AMERICAN SOCIETY OF HEMATOLOGY, & 55TH ANNUAL MEETING OF THE AMERICAN-SOCIETY-OF-HEMATOLOGY; NEW ORLEANS, LA, USA; DECEMBER 07 -10, 2013, vol. 105, no. 8, 1 April 2005 (2005-04-01), & 55TH ANNUAL MEETING OF THE AMERICAN-SOCIETY-OF-HEMATOLOGY; NEW ORLEANS, LA, USA; DECEMBER 07 -10, 2013, pages 3051 - 3057, XP002452476, ISSN: 0006-4971, DOI: 10.1182/blood-2004-07-2974
MIMEAULT, CLIN PHARMACOL THER., vol. 82, no. 3, 1 August 2007 (2007-08-01), pages 252 - 64
MIR, BIOELECTROCHEM, vol. 53, 2000, pages 1 - 10
MORDACQ; LINZER, GENES AND DEV, vol. 3, 1989, pages 760
MOREAU ET AL., NUCL. ACIDS RES., vol. 9, 1981, pages 6047
MUESING ET AL., CELL, vol. 48, 1987, pages 691
NAKAMURA ET AL.: "Handbook of Experimental Immunology (4th Ed.),", vol. 1, 1987, BLACKWELL SCIENTIFIC PUBL, pages: 27
NEUMANN ET AL., PROC. NATL. ACAD. SEI. USA, vol. 96, no. 16, 1999, pages 9345 - 9350
NG ET AL., NUC. ACIDS RES., vol. 17, 1989, pages 601
NOMOTO ET AL., GENE, vol. 236, no. 2, 1999, pages 259 - 271
OMITZ ET AL., MOL. CELL. BIOL.,, vol. 7, 1987, pages 3466
ONDOK ET AL., EMBOJ., vol. 6, 1987, pages 1017
PALMITER ET AL., NATURE, vol. 300, 1982, pages 611
PASSWEG J; TYNDALL A., SEMIN HEMATOL, vol. 4, no. 4, 4 October 2007 (2007-10-04), pages 278 - 85
PASSWEG JR; TICHELLI A; MEYER-MONARD S ET AL.: "Purified donor NK-lymphocytc infusion to consolidate engraftment after haploidentical stem cell transplantation", LEUKEMIA, vol. 18, 2004, pages 1835 - 1838
PECH ET AL., MOL. CELL. BIOL., vol. 9, 1989, pages 396
PELLETIER; SONENBERG, NATURE, vol. 334, no. 6180, 1988, pages 320 - 325
PEREZ-STABLE; CONSTANTINI, MOL. CELL. BIOL., vol. 10, 1990, pages 1116
PICARD; SCHAFFNER, NATURE, vol. 307, 1984, pages 83
PINKERT ET AL., GENES AND DEV., vol. 1, 1987, pages 268
PONTA ET AL., PROC. NATL. ACAD. SEI. USA, vol. 82, 1985, pages 1020
PORTON ET AL., MOL. CELL. BIOL., vol. 10, 1990, pages 1076
QUEEN; BALTIMORE, CELL, vol. 35, 1983, pages 741
QUINN ET AL., MOL. CELL. BIOL., vol. 9, 1989, pages 4713
RABINOVICH PETER M ET AL: "Synthetic messenger RNA as a tool for gene therapy.", HUMAN GENE THERAPY, LIEBERT, US, vol. 17, no. 10, 1 October 2006 (2006-10-01), US, pages 1027 - 1035, XP002489890, ISSN: 1043-0342, DOI: 10.1089/hum.2006.17.1027
REDONDO ET AL., SCIENCE, vol. 247, 1990, pages 1225
REISMAN; ROTTER, MOL. CELL. BIOL., vol. 9, 1989, pages 3571
RESENDEZ JR., MOL. CELL. BIOL., vol. 8, 1988, pages 4579
RIPE ET AL., MOL. CELL. BIOL., vol. 9, 1989, pages 2224
RITTLING ET AL., NUC. ACIDSRES., vol. 17, 1989, pages 1619
ROLS; TEISSIE, BIOPHYS. J.,, vol. 75, no. 3, 1998, pages 1415 - 1423
ROSEN ET AL., CELL, vol. 41, 1988, pages 813
RUGGERI L, ET AL: "Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants", SCIENCE, AAAS, AMERICAN ASSOC. FOR THE ADVANCEMENT OF SCIENCE, US, vol. 295, no. 5562, 15 March 2002 (2002-03-15), US, pages 2097 - 2100, XP002382437, ISSN: 0036-8075, DOI: 10.1126/science.1068440
RUGGERI LOREDANA; MANCUSI ANTONELLA; BURCHIELLI EMANUELA; AVERSA FRANCO; MARTELLI MASSIMO F; VELARDI ANDREA: "Natural killer cell alloreactivity in allogeneic hematopoietic transplantation", CURRENT OPINION IN ONCOLOGY, LIPPINCOTT WILLIAMS & WILKINS, LTD., GB, vol. 19, no. 2, 1 March 2007 (2007-03-01), GB, pages 142 - 147, XP009122006, ISSN: 1040-8746, DOI: 10.1097/CCO.0b013e3280148a1a
SAKAI ET AL., GENES AND DEV., vol. 2, 1988, pages 1144
SAMBROOK ET AL.: "Molecular cloning", 2001, COLD SPRING HARBOR LABORATORY PRESS
SATAKE ET AL., J. VIROLOGY, vol. 62, 1988, pages 970
SCHAFFNER ET AL., J. MOL. BIOL., vol. 201, 1988, pages 81
SEARLE ET AL., MOL. CELL. BIOL., vol. 5, 1985, pages 1480
See also references of EP2279253A4
SHARP; MARCINIAK, CELL, vol. 59, 1989, pages 229
SHAUL; BEN-LEVY, EMBOJ., vol. 6, 1987, pages 1913
SHERMAN ET AL., MOL. CELL. BIOL., vol. 9, 1989, pages 50
SLEIGH; LOCKETT, J. EMBO, vol. 4, 1985, pages 3831
SPALHOLZ ET AL., CELL, vol. 42, 1985, pages 183
SPANDAU; LEE, J. VIROLOGY, vol. 62, 1988, pages 427
SPANDIDOS; WILKIE, EMBOJ., vol. 2, 1983, pages 1193
STEPHENS; HENTSCHEL, BIOCHEM. J., vol. 248, 1987, pages 1
STRENGALL ET AL., J IMMUNOLOGY, vol. 170, 2003, pages 5464 - 5469
STUART ET AL., NATURE, vol. 317, 1985, pages 828
SULLIVAN; PETERLIN, MOL. CELL. BIOL., vol. 7, 1987, pages 3315
SWARTZENDRUBER; AND LEHMAN, J. CELL. PHYSIOLOGY, vol. 85, 1975, pages 179
TAKEBE ET AL., MOL. CELL. BIOL., vol. 8, 1988, pages 466
TAVEMIER ET AL., NATURE, vol. 301, 1983, pages 634
TAYLOR ET AL., J. BIOL. CHEM., vol. 264, 1989, pages 15160
TAYLOR; KINGSTON, MOL. CELL. BIOL., vol. 10, 1990, pages 165
TAYLOR; KINGSTON, MOL. CELL. BIOL., vol. 10, 1990, pages 176
TEUFEL ET AL., CELL. MOL. LIFE SCI, vol. 62, 2005, pages 1755 - 1762
THIESEN ET AL., J. VIROLOGY, vol. 62, 1988, pages 614
TREISMAN, CELL, vol. 42, 1985, pages 889
TROMPETER, H.-I. WEINHOLD, S. THIEL, C. WERNET, P. UHRBERG, M.: "Rapid and highly efficient gene transfer into natural killer cells by nucleofection", JOURNAL OF IMMUNOLOGICAL METHODS., ELSEVIER SCIENCE PUBLISHERS B.V.,AMSTERDAM., NL, vol. 274, no. 1-2, 1 March 2003 (2003-03-01), NL, pages 245 - 256, XP004411931, ISSN: 0022-1759, DOI: 10.1016/S0022-1759(02)00431-3
TRONCHE ET AL., MOL. BIOL. MED., vol. 7, 1990, pages 173
TRUDEL; CONSTANTINI, GENES AND DEV, vol. 6, 1987, pages 954
TSUMAKI ET AL., J. BIOL. CHERN.,, vol. 273, no. 36, 1998, pages 22861 - 22864
TYNDELL ET AL., NUC. ACIDS. RES., vol. 9, 1981, pages 6231
VAN DE PARRE, T.J.L. MARTINET, W. SCHRIJVERS, D.M. HERMAN, A.G. DE MEYER, G.R.Y.: "mRNA but not plasmid DNA is efficiently transfected in murine J774A.1 macrophages", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, ELSEVIER, AMSTERDAM, NL, vol. 327, no. 1, 4 February 2005 (2005-02-04), AMSTERDAM, NL, pages 356 - 360, XP004697935, ISSN: 0006-291X, DOI: 10.1016/j.bbrc.2004.12.027
VAN MEIRVENNE ET AL., CANCER GENE THER., vol. 9, no. 9, 2002, pages 787 - 797
VAN SCHOOTEN ET AL., MOL MED TODAY, vol. 3, no. 6, 1997, pages 254 - 260
VAN TENDELOO ET AL., BLOOD, vol. 98, no. I, 2001, pages 49 - 56
VANNICE; LEVINSON, J. VIROLOGY, vol. 62, 1988, pages 1305
VASSEUR ET AL., PROC NATL. ACAD. SEI. U.S.A., vol. 77, 1980, pages 1068
WANG; CALAME, CELL, vol. 47, 1986, pages 241
WARD ET AL., CATHETER CARDIOVASC INTERV., vol. 70, no. 7, 1 December 2007 (2007-12-01), pages 983 - 98
WEAVER; CHIZMADZHEV, BIOENERG., vol. 41, 1996, pages 135 - 160
WEBER ET AL., CELL, vol. 36, 1984, pages 983
WEINBERGER ET AL., MOL. CELL. BIOL., vol. 8, 1984, pages 988
WEISS ET AL., PROC. AM. ASSOC. CANCER RES., vol. 44, 2003, pages 1094 - 1095
WEISS ET AL., PROC. AM. ASSOC. CANCER RES., vol. 44, no. 5493, 2003, pages 1094 - 1095
WENDTNER; LEUK, LYMPHOMA, vol. 45, no. 5, 2004, pages 897 - 904
WIERDA ET AL., BLOOD, vol. 96, no. 9, 2000, pages 2917 - 2924
WINOTO; BALTIMORE, CELL, vol. 59, 1989, pages 649
WU ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 233, no. 1, 1997, pages 221 - 2266
XIE TD; SUN F; TSONG TY: "Study of mechanisms of electric field-induced DNA transfection. I. DNA entry by surface binding and diffusion through membrane pores", BIOPHYSJ., vol. 58, 1990, pages 13 - 19
Y ZHAO, Z ZHENG, C COHEN, L GATTINONI, D PALMER, N RESTIFO, S ROSENBERG, R MORGAN: "High-Efficiency Transfection of Primary Human and Mouse T Lymphocytes Using RNA Electroporation", MOLECULAR THERAPY, NATURE PUBLISHING GROUP, GB, vol. 13, no. 1, 1 January 2006 (2006-01-01), GB, pages 151 - 159, XP055267774, ISSN: 1525-0016, DOI: 10.1016/j.ymthe.2005.07.688
YUTZEY ET AL., MOL. CELL. BIOL., vol. 9, 1989, pages 1397
ZHAO-EMONET ET AL., BIOCHIM. BIOPHYS. ACTA, vol. 1442, no. 2-3, 1998, pages 109 - 119
ZHOU ET AL., J. BIOL. CHEM., vol. 270, no. 21, 1995, pages 12665 - 12669
ZIMMERMANN U.: "Electrical breakdown, electropermeabilization and electrofusion", REV PHYSIOL BIOCHEM PHARMACOL, vol. 1, no. 05, 1986, pages 176 - 256

Also Published As

Publication number Publication date
JP2017184759A (en) 2017-10-12
US20180028567A1 (en) 2018-02-01
US11331344B2 (en) 2022-05-17
US20090257991A1 (en) 2009-10-15
US9669058B2 (en) 2017-06-06
US10660917B2 (en) 2020-05-26
US20170258837A1 (en) 2017-09-14
US20200330519A1 (en) 2020-10-22
US9132153B2 (en) 2015-09-15
JP2011517944A (en) 2011-06-23
DK2279253T3 (en) 2017-02-13
JP6630074B2 (en) 2020-01-15
US20160082045A1 (en) 2016-03-24
EP2279253A4 (en) 2011-12-21
JP5779090B2 (en) 2015-09-16
US20220265722A1 (en) 2022-08-25
US20200237825A1 (en) 2020-07-30
EP2279253A2 (en) 2011-02-02
JP2016005459A (en) 2016-01-14
EP2279253B1 (en) 2016-11-16
JP2020012000A (en) 2020-01-23
US20140017213A1 (en) 2014-01-16
WO2009126789A3 (en) 2010-01-28
US8450112B2 (en) 2013-05-28
JP6944497B2 (en) 2021-10-06

Similar Documents

Publication Publication Date Title
US20220265722A1 (en) Engineering and delivery of therapeutic compositions of freshley isolated cells
US20240115609A1 (en) METHODS FOR EXPANDING AND ACTIVATING yo T CELLS FOR THE TREATMENT OF CANCER AND RELATED MALIGNANCIES
US11766456B2 (en) Method for culturing natural killer cells using T cells
EP2700708B1 (en) Enhancing the T-cell stimulatory capacity of human antigen presenting cells in vitro and in vivo and its use in vaccination
JP2022065022A (en) Methods for generating engineered human primary blood dendritic cell lines
US20170000881A1 (en) Enhancing the t-cell stimulatory capacity of human antigen presenting cells in vitro and in vivo and its use in vaccination
AU2020213119A1 (en) Compositions and methods for targeting mutant RAS
EP3730515A1 (en) A fully-human t cell receptor specific for the 369-377 epitope derived from the her2/neu (erbb2) receptor protein
Wu et al. Extracellular domain of human 4-1BBL enhanced the function of cytotoxic T-lymphocyte induced by dendritic cell

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09731422

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2011504171

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2009731422

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2009731422

Country of ref document: EP